1
|
Dave RH, Crayton H, Miravalle A, Tai MH, Wyse K, Houghton K, Hitchens A, Berkovich R. COVID-19 Vaccination Response in Patients with Multiple Sclerosis Treated with Ofatumumab in the United States: A Medical Record Review. Neurol Ther 2024:10.1007/s40120-024-00671-0. [PMID: 39441497 DOI: 10.1007/s40120-024-00671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Real-world data are required to provide a greater understanding of the impact of ofatumumab on the ability to mount an effective immune response following the receipt of approved COVID-19 vaccinations. This retrospective real-world analysis aimed to describe the humoral immune response to COVID-19 vaccination during ofatumumab treatment in patients with multiple sclerosis (MS). METHODS Data from patients with MS treated with ofatumumab who were fully vaccinated against COVID-19 infection were abstracted from medical charts at four clinical sites in the USA. Patient characteristics and humoral response were summarized descriptively. Differences in humoral response were documented on the basis of vaccination status during ofatumumab treatment (i.e., after full vaccination and after booster vaccination) and prior disease-modifying treatment (DMT) exposure (i.e., DMT naïve, prior anti-CD20/sphingosine 1-phosphate [S1P] therapy, prior non-anti-CD20/S1P therapy). The sample size precluded formal statistical analysis. RESULTS Thirty-eight patients were included. The mean (standard deviation) duration of ofatumumab treatment upon data collection was 20.4 (4.6) months (treatment ongoing for 35 [92%] patients). Definitive humoral response after full vaccination was documented for 34 patients, of whom 20 (60%) were seropositive. Definitive humoral response after booster vaccination was documented among five patients, of whom three (60%) were seropositive. Among patients who were DMT naïve prior to ofatumumab (n = 15), 73% were seropositive; among patients exposed to prior anti-CD20/S1P therapy (n = 14), 33% were seropositive; and among patients exposed to prior non-anti-CD20/S1P therapy (n = 9), 56% were seropositive. Patients naïve to DMT had been living with an MS diagnosis for a shorter duration than those experienced with DMTs. CONCLUSION Patients with MS receiving ongoing treatment with ofatumumab can mount a positive humoral response to a COVID-19 vaccination. Prior treatment with anti-CD20 or S1P DMTs may be a risk factor for lower humoral response.
Collapse
Affiliation(s)
- Rahul H Dave
- Inova Medical Group-Neurology II, Fairfax, VA, USA
| | - Heidi Crayton
- Multiple Sclerosis Center of Greater Washington, Washington, VA, USA
| | | | - Ming-Hui Tai
- Novartis Pharmaceuticals Corporation, One Health Plaza East, Hanover, NJ, 07936, USA.
| | - Kerri Wyse
- Novartis Pharmaceuticals Corporation, One Health Plaza East, Hanover, NJ, 07936, USA
| | | | | | - Regina Berkovich
- Los Angeles County General Hospital and Clinic, Los Angeles, CA, USA
| |
Collapse
|
2
|
Torgauten HM, Onyango TB, Ljostveit S, Hallin EI, Serkland TT, Skrede S, Langeland N, Cox RJ, Wergeland S, Myhr KM, Torkildsen Ø. Hospitalisations and humoral COVID-19 vaccine response in vaccinated rituximab-treated multiple sclerosis patients. Mult Scler Relat Disord 2024; 89:105770. [PMID: 39029342 DOI: 10.1016/j.msard.2024.105770] [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: 01/02/2024] [Revised: 06/07/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Patients with multiple sclerosis (MS) treated with anti-CD20 therapies such as rituximab may have increased risk of severe COVID-19 disease. Vaccination induces protective immunity, but humoral vaccine response is known to be attenuated in rituximab-treated MS-patients-patients, which has indicated a need for real world data on severe morbidity and mortality from COVID-19 after vaccination. METHODS Rituximab-treated patients treated at Haukeland University Hospital were identified through the National MS Registry and invited to participate in the study by giving a consent and providing a blood sample 3 weeks or later after ordinary COVID-19- vaccination, i.e. 2 doses given with a standard interval of 3 weeks. Blood samples were analysed with Enzyme-Linked Immunosorbent assay (ELISA) to evaluate humoral vaccine response with screening test against receptor-binding domain (RBD) and confirmatory Spike IgG-specific ELISA. A haemagglutination test (HAT) was performed as a marker of neutralizing antibodies. Patient serum concentration of rituximab were quantified using liquid chromatography tandem mass spectrometry (LC-MS/MS). Registry data from the Norwegian MS registry and information on hospitalization from patient records were collected and linked to laboratory results. RESULTS 111 patients were included in the study. A total of 7 (6.3%) were hospitalized due to COVID-19 disease during the observation period. No patient was admitted to ICU and there were no deaths. 34.2% did not have detectable titre of SARS CoV-2 Spike IgG antibodies, 72.1% did not have a detectable titre of SARS CoV-2 RBD antibodies, and 88.2% did not have a detectable HAT titre. There was a correlation between hospitalisation and the absence of SARS CoV-2 Spike IgG antibody titre, and between hospitalisation and MS disease duration, as well as between spike IgG antibody titre and CD19 B-cell count, time since last rituximab infusion, cumulative rituximab treatment time and total IgG level in the patients. CONCLUSION A substantial proportion of rituximab-treated MS-patients-patients did not have detectable humoral vaccine responses after 2 doses of COVID-19 vaccination. Despite this, the cumulative percentage of patients hospitalized with COVID-19 disease throughout the observation period of 22 months was low, and no patients required ICU treatment. The results support that vaccinated MS-patients treated with rituximab have a protective effect against serious Covid-19 infection.
Collapse
Affiliation(s)
- Hilde M Torgauten
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Sonja Ljostveit
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Erik I Hallin
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Trond T Serkland
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Silje Skrede
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, Bergen, Norway; National Advisory Unit for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian MS Registry, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Kjell-Morten Myhr
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Øivind Torkildsen
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
3
|
Wüsthoff LEC, Lund-Johansen F, Henriksen K, Wildendahl G, Jacobsen JA, Gomes L, Anjum HS, Barlinn R, Kran AMB, Munthe LA, Vaage JT. Seroprevalence of SARS-CoV-2 and humoral immune responses to COVID-19 mRNA vaccines among people who use drugs - in the light of tailored mitigating strategies. Harm Reduct J 2024; 21:120. [PMID: 38890611 PMCID: PMC11186241 DOI: 10.1186/s12954-024-01023-9] [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: 05/16/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND During the initial wave of the COVID-19 pandemic, there was a surprisingly low incidence of SARS-CoV-2 among People Who Use Drugs (PWUD) in Oslo, Norway, despite their heightened vulnerability regarding risk of infection and severe courses of the disease.This study aims to investigate the seroprevalence of SARS-CoV-2 antibodies among PWUD, their antibody responses to relevant virus infections and COVID-19 mRNA vaccines, and their vaccination coverage compared to the general population. METHODS Conducted as a prospective cohort study, data was collected from residents in six institutions for homeless PWUD and users of a low-threshold clinic for opioid agonist treatment. Ninety-seven participants were recruited for SARS-CoV-2 seroprevalence analysis. Additional two participants with known positive SARS-CoV-2 test results were recruited for further analyses. Twenty-five participants completed follow-up. Data included questionnaires, nasal swabs and blood samples. Data on vaccination coverage was obtained from the National Vaccine Register. Serologic methods included detection of antibodies to relevant virus proteins, neutralizing antibodies to SARS-CoV-2, antibodies to the full-length spike protein, and receptor-binding domain from SARS-CoV-2. RESULTS Among PWUD, antibodies to SARS-CoV-2 were detected in 2 out of 97 samples before vaccines against SARS-CoV-2 were available, comparable to a 2.8% frequency in population-based screening. Levels of serum antibodies to seasonal coronaviruses and Epstein-Barr-Virus (EBV) in PWUD were similar to population-based levels. After the second vaccine dose, binding and neutralizing antibody levels to SARS-CoV-2 in PWUD were comparable to controls. Eighty-four of PWUD received at least one dose of COVID-19 mRNA vaccine, compared to 89% in the general population. CONCLUSION Results indicate that PWUD did not exhibit increased SARS-CoV-2 seroprevalence or elevated serum antibodies to seasonal coronaviruses and EBV. Moreover, vaccine responses in PWUD were comparable to controls, suggesting that vaccination is effective in conferring protection against SARS-CoV-2 also in this population.
Collapse
Affiliation(s)
- Linda Elise Couëssurel Wüsthoff
- Unit for Clinical Research on Addictions, Oslo University Hospital, PO Box 4959 Nydalen, Oslo, 0424, Norway.
- Norwegian Centre for Addiction Reasearch, Institute of Clinical Medicine, University of Oslo, PO Box 1039 Blindern, Oslo, 0315, Norway.
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Gaustadalleen 21, Oslo, 0349, Norway
- Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, Oslo, 0318, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, 0318, Oslo, Norway
| | - Kathleen Henriksen
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
- Student Health Services, Student Welfare Services in Oslo, Problemveien 9, Oslo, 0313, Norway
| | - Gull Wildendahl
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Jon-Aksel Jacobsen
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Leni Gomes
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Hina Sarwar Anjum
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Regine Barlinn
- Department of Microbiology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
| | - Anne-Marte Bakken Kran
- Division of Infection Control, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo, 0213, Norway
| | - Ludvig Andre Munthe
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- KG Jebsen Centre for B cell Malignancies, and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, PO Box 4950 Nydalen, Oslo, 0424, Norway
| | - John T Vaage
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, Oslo, 0318, Norway
| |
Collapse
|
4
|
Gualtieri R, Yerly S, Garcia-Tarodo S, Parvex P, Rock N, Posfay-Barbe K, Didierlaurent AM, Eberhardt C, Blanchard-Rohner G. Rituximab-to-vaccine interval on SARS-CoV-2 immunogenicity in children: The potential role of prior natural infection. Pediatr Allergy Immunol 2024; 35:e14161. [PMID: 38796784 DOI: 10.1111/pai.14161] [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: 12/22/2023] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Treatment with anti-CD20 antibodies (rituximab) is used in both adults and children to treat various autoimmune and oncological diseases. Rituximab depletes B CD20+ cells and, thereby, antibody response to vaccines. This study aimed to examine the antibody response to mRNA-based COVID-19 vaccines in children aged 5-18 years undergoing rituximab treatment compared to healthy matched children. METHODS Between 31 January and 18 July 2022, we conducted a prospective observational study at the Geneva University Hospitals, enrolling children aged 5-18 years under rituximab treatment who had received two mRNA-based SARS-CoV-2 vaccine doses. Controls were healthy volunteers with no significant medical conditions. Exclusion criteria included a recent SARS-CoV-2 infection. Blood samples were collected at day 60 (±30) and day 270 (±90) after the second vaccination. RESULTS The rituximab-treated group exhibited significantly lower levels of antibodies specific to the anti-receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein than healthy controls at 60 (±30) days after the second vaccine dose (geometric mean concentration: 868.3 IU/mL in patients and 11,393 IU/mL in controls; p = .008). However, patients with a rituximab-to-vaccine interval shorter than 6 months and with evidence of a past infection (based on positive anti-N antibody levels) had a high level of anti-RBD antibodies. CONCLUSION A past infection with SARS-CoV-2 may induce anti-RBD-specific memory B cells that can be re-activated by SARS-CoV-2 vaccination, even after rituximab-induced B-cell depletion. This suggests that it is possible to vaccinate earlier than 6 months after rituximab to develop a good antibody response, especially in the case of past SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Renato Gualtieri
- Pediatric Platform for Clinical Research, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sabine Yerly
- Laboratory of Virology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Stephanie Garcia-Tarodo
- Unit of Pediatric Neurology, Division of Pediatric Specialties, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| | - Paloma Parvex
- Unit of Pediatric Nephrology, Division of Pediatric Specialties, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| | - Nathalie Rock
- Pediatric Gastroenterology, Hepatology and Nutrition Unit, Department of Pediatrics, Gynecology and Obstetrics, Swiss Pediatric Liver Center, Geneva University Hospitals, Geneva, Switzerland
- Department of Pediatrics, Gynecology, and Obstetrics, Swiss Pediatric Liver Center, University of Geneva, University Hospitals of Geneva, Geneva, Switzerland
| | - Klara Posfay-Barbe
- Pediatric Platform for Clinical Research, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Unit of Infectious Diseases, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| | - Arnaud M Didierlaurent
- Department of Pathology and Immunology, Center for Vaccinology, University of Geneva, Geneva, Switzerland
| | - Christiane Eberhardt
- Department of Pathology and Immunology, Center for Vaccinology, University of Geneva, Geneva, Switzerland
- Center for Vaccinology, University Hospitals of Geneva, Geneva, Switzerland
| | - Geraldine Blanchard-Rohner
- Unit of Immunology, Vaccinology and Rheumatology, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
5
|
Ju H, Seok JM, Chung YH, Jeon MY, Lee HL, Kwon S, Kim S, Min JH, Kim BJ. Evaluation of SARS-CoV-2 Vaccine-Induced Antibody Responses in Patients with Neuroimmunological Disorders: A Real-World Experience. Diagnostics (Basel) 2024; 14:502. [PMID: 38472974 DOI: 10.3390/diagnostics14050502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
This study evaluates the antibody responses to SARS-CoV-2 vaccines in patients with neuroimmunological disorders (pwNID) who are receiving immunomodulating treatments, compared to healthy individuals. It included 25 pwNID with conditions such as optic neuritis, neuromyelitis optica spectrum disorder, multiple sclerosis, myasthenia gravis, and polymyositis, as well as 56 healthy controls. All participants had completed their full SARS-CoV-2 vaccination schedule, and their blood samples were collected within six months of their last dose. The concentration of anti-SARS-CoV-2 IgG antibodies was measured using an enzyme-linked immunosorbent assay. The results showed that pwNID had significantly lower antibody titers (58.4 ± 49.2 RU/mL) compared to healthy individuals (81.7 ± 47.3 RU/mL). This disparity persisted even after adjusting for age and the interval between the final vaccination and sample collection. A notable correlation was found between the use of immunomodulating treatments and reduced antibody levels, whereas mRNA vaccines were linked to higher antibody concentrations. The conclusion of this study is that immunomodulating treatments may reduce the effectiveness of SARS-CoV-2 vaccines in pwNID. This insight is crucial for healthcare providers in designing vaccination strategies and managing treatment plans for pwNID on immunomodulating therapies, highlighting the need for personalized approaches in this subgroup.
Collapse
Affiliation(s)
- Hyunjin Ju
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Neurology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| | - Jin Myoung Seok
- Department of Neurology, Soonchunhyang University Hospital Cheonan, Soonchunhyang University College of Medicine, Cheonan 31193, Republic of Korea
| | - Yeon Hak Chung
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Mi Young Jeon
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hye Lim Lee
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Soonwook Kwon
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Sunyoung Kim
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Ju-Hong Min
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Byoung Joon Kim
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
6
|
Millán-Pascual J, Valero-López G, Iniesta-Martinez F, Hellin-Gil MF, Jimenez-Veiga J, López-Tovar IA, Morales-Ortiz A, Meca-Lallana JE. Humoral Response to SARS-COV-2 Vaccination in Patients with Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder: A Real-World Study. Neurol Ther 2024; 13:153-164. [PMID: 38097868 PMCID: PMC10787726 DOI: 10.1007/s40120-023-00572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/01/2023] [Indexed: 01/14/2024] Open
Abstract
INTRODUCTION The risk of SARS-CoV-2 infection or severe coronavirus disease 2019 (COVID-19) has been shown to increase in patients with multiple sclerosis (MS). Vaccination is recommended in this patient population, and the effect of disease-modifying treatments (DMTs) on response to vaccination should be considered. METHODS This prospective, observational, cross-sectional study investigated humoral response after COVID-19 vaccination as well as possible predictors for response in patients with MS and other neuroinflammatory diseases who received DMTs in routine clinical practice in Spain. Responses were compared versus those seen in healthy controls. RESULTS After vaccination against COVID-19, most patients with MS developed an immune response comparable to that of healthy individuals. However, approximately half of patients receiving a sphingosine-1-phosphate modulator (SP1-M, fingolimod or siponimod) or a B-cell-depleting agent (aCD20, ocrelizumab or rituximab) did not develop protective antibodies, although patients receiving other DMTs had humoral immune responses comparable to healthy controls. Lymphocyte count was not associated with reduced humoral response in patients receiving an SP1-M or aCD20, whereas, in patients receiving an aCD20 or SP1-M, older age was associated with lower anti-SARS-CoV-2 spike protein immunoglobulin G antibody levels. CONCLUSIONS Treatment with aCD20 or SP1-M therapies appears to be associated with a lower humoral response to vaccines against SARS-CoV-2. Vaccination prior to initiation of these DMTs should be recommended whenever possible.
Collapse
Affiliation(s)
- Jorge Millán-Pascual
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain.
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain.
| | - Gabriel Valero-López
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain
| | - Francisca Iniesta-Martinez
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain
| | - Maria Fuensanta Hellin-Gil
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain
| | - Judith Jimenez-Veiga
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain
| | | | - Ana Morales-Ortiz
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
| | - José E Meca-Lallana
- Multiple Sclerosis CSUR and Clinical Neuroimmunology Unit, Neurology Department, Virgen de la Arrixaca University Hospital (IMIB-Arrixaca), Murcia, Spain
- Clinical Neuroimmunology and Multiple Sclerosis Cathedra, UCAM, Universidad Católica San Antonio, Murcia, Spain
| |
Collapse
|
7
|
Marantos T, Kyriazopoulou E, Lekakis V, Voumvourakis KI, Tsiodras S. Immunogenicity and safety of vaccines in multiple sclerosis: A systematic review and meta-analysis. J Neurol Sci 2024; 456:122852. [PMID: 38142541 DOI: 10.1016/j.jns.2023.122852] [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: 10/12/2023] [Revised: 12/06/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Seroconversion rate of vaccines varies and requires further elucidation in patients with multiple sclerosis (MS) under treatment with disease-modifying therapies (DMTs). We aimed to investigate this in a systematic review and meta-analysis. METHODS MEDLINE(PubMed) and Cochrane databases were searched based on a pre-specified protocol (PROSPERO: CRD42020202018). Studies reporting on patients with MS, diagnosed with McDonald criteria getting vaccinated with any type of vaccine were included in the analysis. The primary endpoint was the incidence of patients being seropositive and experience adverse events after vaccination. Outcomes were expressed as proportions with respective 95% confidence interval (CI). Two reviewers independently screened and reviewed existing literature and assessed study quality with the Methodological index for non-randomized studies. RESULTS Of 295 articles, 45 studies were analyzed. Seroconversion after COVID-19 vaccines was 76% (95% CI, 70-80; I2 = 95%; 20 studies including 5601 patients. Protection was lower in patients treated with anti-CD20 antibodies and sphingosine-1-phosphate receptor (S1PR) modulators compared to untreated patients or treatment with other DMTs. Relapse occurred in 2% (95% CI, 1-3; I2 = 86%; 16 studies including 7235 patients). Seroconversion after seasonal influenza vaccines was 82% (95% CI, 65-91; I2 = 90%; 6 studies including 490 patients). Relapse rate was similar to this after COVID-19 vaccination. CONCLUSION The majority of MS patients vaccinated for COVID-19 or seasonal influenza mount an adequate immune response without safety concerns. Data on other vaccines are limited.
Collapse
Affiliation(s)
- Theodoros Marantos
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Evdoxia Kyriazopoulou
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Vasileios Lekakis
- Department of Gastroenterology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Sotirios Tsiodras
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| |
Collapse
|
8
|
Benjakul S, Anthi AK, Kolderup A, Vaysburd M, Lode HE, Mallery D, Fossum E, Vikse EL, Albecka A, Ianevski A, Kainov D, Karlsen KF, Sakya SA, Nyquist-Andersen M, Gjølberg TT, Moe MC, Bjørås M, Sandlie I, James LC, Andersen JT. A pan-SARS-CoV-2-specific soluble angiotensin-converting enzyme 2-albumin fusion engineered for enhanced plasma half-life and needle-free mucosal delivery. PNAS NEXUS 2023; 2:pgad403. [PMID: 38077689 PMCID: PMC10703496 DOI: 10.1093/pnasnexus/pgad403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/13/2023] [Indexed: 02/29/2024]
Abstract
Immunocompromised patients often fail to raise protective vaccine-induced immunity against the global emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Although monoclonal antibodies have been authorized for clinical use, most have lost their ability to potently neutralize the evolving Omicron subvariants. Thus, there is an urgent need for treatment strategies that can provide protection against these and emerging SARS-CoV-2 variants to prevent the development of severe coronavirus disease 2019. Here, we report on the design and characterization of a long-acting viral entry-blocking angiotensin-converting enzyme 2 (ACE2) dimeric fusion molecule. Specifically, a soluble truncated human dimeric ACE2 variant, engineered for improved binding to the receptor-binding domain of SARS-CoV-2, was fused with human albumin tailored for favorable engagement of the neonatal fragment crystallizable receptor (FcRn), which resulted in enhanced plasma half-life and allowed for needle-free transmucosal delivery upon nasal administration in human FcRn-expressing transgenic mice. Importantly, the dimeric ACE2-fused albumin demonstrated potent neutralization of SARS-CoV-2 immune escape variants.
Collapse
Affiliation(s)
- Sopisa Benjakul
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Aina Karen Anthi
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Anette Kolderup
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Marina Vaysburd
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Heidrun Elisabeth Lode
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Donna Mallery
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Even Fossum
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Elisabeth Lea Vikse
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Anna Albecka
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
- Institute of Technology, University of Tartu, Tartu 50411, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00290, Finland
| | - Karine Flem Karlsen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
| | - Siri Aastedatter Sakya
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Mari Nyquist-Andersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Torleif Tollefsrud Gjølberg
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Morten C Moe
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Magnar Bjørås
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo 0371, Norway
| | - Leo C James
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Jan Terje Andersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| |
Collapse
|
9
|
Asashima H, Kim D, Wang K, Lele N, Buitrago-Pocasangre NC, Lutz R, Cruz I, Raddassi K, Ruff WE, Racke MK, Wilson JE, Givens TS, Grifoni A, Weiskopf D, Sette A, Kleinstein SH, Montgomery RR, Shaw AC, Li F, Fan R, Hafler DA, Tomayko MM, Longbrake EE. Prior cycles of anti-CD20 antibodies affect antibody responses after repeated SARS-CoV-2 mRNA vaccination. JCI Insight 2023; 8:e168102. [PMID: 37606046 PMCID: PMC10543713 DOI: 10.1172/jci.insight.168102] [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: 12/15/2022] [Accepted: 07/06/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUNDWhile B cell depletion is associated with attenuated antibody responses to SARS-CoV-2 mRNA vaccination, responses vary among individuals. Thus, elucidating the factors that affect immune responses after repeated vaccination is an important clinical need.METHODSWe evaluated the quality and magnitude of the T cell, B cell, antibody, and cytokine responses to a third dose of BNT162b2 or mRNA-1273 mRNA vaccine in patients with B cell depletion.RESULTSIn contrast with control individuals (n = 10), most patients on anti-CD20 therapy (n = 48) did not demonstrate an increase in spike-specific B cells or antibodies after a third dose of vaccine. A third vaccine elicited significantly increased frequencies of spike-specific non-naive T cells. A small subset of B cell-depleted individuals effectively produced spike-specific antibodies, and logistic regression models identified time since last anti-CD20 treatment and lower cumulative exposure to anti-CD20 mAbs as predictors of those having a serologic response. B cell-depleted patients who mounted an antibody response to 3 vaccine doses had persistent humoral immunity 6 months later.CONCLUSIONThese results demonstrate that serial vaccination strategies can be effective for a subset of B cell-depleted patients.FUNDINGThe NIH (R25 NS079193, P01 AI073748, U24 AI11867, R01 AI22220, UM 1HG009390, P01 AI039671, P50 CA121974, R01 CA227473, U01CA260507, 75N93019C00065, K24 AG042489), NIH HIPC Consortium (U19 AI089992), the National Multiple Sclerosis Society (CA 1061-A-18, RG-1802-30153), the Nancy Taylor Foundation for Chronic Diseases, Erase MS, and the Claude D. Pepper Older Americans Independence Center at Yale (P30 AG21342).
Collapse
Affiliation(s)
- Hiromitsu Asashima
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kaicheng Wang
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Nikhil Lele
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Rachel Lutz
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Isabella Cruz
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Khadir Raddassi
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - William E. Ruff
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Repertoire Immune Medicines, Cambridge, Massachusetts, USA
| | | | | | | | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, UCSD, La Jolla, California, USA
| | - Steven H. Kleinstein
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| | | | - Albert C. Shaw
- Section of Infectious Diseases, Department of Internal Medicine, and
| | - Fangyong Li
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - David A. Hafler
- Department of Neurology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mary M. Tomayko
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
10
|
Otero-Romero S, Lebrun-Frénay C, Reyes S, Amato MP, Campins M, Farez M, Filippi M, Hacohen Y, Hemmer B, Juuti R, Magyari M, Oreja-Guevara C, Siva A, Vukusic S, Tintoré M. ECTRIMS/EAN consensus on vaccination in people with multiple sclerosis: Improving immunization strategies in the era of highly active immunotherapeutic drugs. Mult Scler 2023; 29:904-925. [PMID: 37293841 PMCID: PMC10338708 DOI: 10.1177/13524585231168043] [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: 11/04/2022] [Revised: 01/30/2023] [Accepted: 03/19/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND With the new highly active drugs available for people with multiple sclerosis (pwMS), vaccination becomes an essential part of the risk management strategy. OBJECTIVE To develop a European evidence-based consensus for the vaccination strategy of pwMS who are candidates for disease-modifying therapies (DMTs). METHODS This work was conducted by a multidisciplinary working group using formal consensus methodology. Clinical questions (defined as population, interventions, and outcomes) considered all authorized DMTs and vaccines. A systematic literature search was conducted and quality of evidence was defined according to the Oxford Centre for Evidence-Based Medicine Levels of Evidence. The recommendations were formulated based on the quality of evidence and the risk-benefit balance. RESULTS Seven questions, encompassing vaccine safety, vaccine effectiveness, global vaccination strategy and vaccination in sub-populations (pediatric, pregnant women, elderly and international travelers) were considered. A narrative description of the evidence considering published studies, guidelines, and position statements is presented. A total of 53 recommendations were agreed by the working group after three rounds of consensus. CONCLUSION This first European consensus on vaccination in pwMS proposes the best vaccination strategy according to current evidence and expert knowledge, with the goal of homogenizing the immunization practices in pwMS.
Collapse
Affiliation(s)
- Susana Otero-Romero
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | | | - Saúl Reyes
- Fundación Santa Fe de Bogotá, Bogotá, Colombia School of Medicine, Universidad de los Andes, Bogotá, Colombia Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Maria Pia Amato
- Department NEUROFARBA, University of Florence, Florence, Italy IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Magda Campins
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | - Mauricio Farez
- Centro para la Investigación de Enfermedades Neuroinmunológicas (CIEN), FLENI, Buenos Aires, Argentina
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy Neurology Unit, Neurorehabilitation Unit, and Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Yael Hacohen
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, UK Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Rosa Juuti
- Multiple Sclerosis International Federation, London, UK
| | - Melinda Magyari
- Department of Neurology, Danish Multiple Sclerosis Center and the Danish Multiple Sclerosis Registry, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, IdISSC, Departamento de Medicina, Universidad Complutense, Madrid, Spain
| | - Aksel Siva
- Department of Neurology, School of Medicine, Istanbul University Cerrahpasa, Cerrahpasa, Istanbul, Turkey
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
- Centre des Neurosciences de Lyon, Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Mar Tintoré
- Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| |
Collapse
|
11
|
Seree-aphinan C, Ratanapokasatit Y, Suchonwanit P, Rattanakaemakorn P, O-Charoen P, Pisitkun P, Suangtamai T, Setthaudom C, Chirasuthat S, Chanprapaph K. Optimal time for COVID-19 vaccination in rituximab-treated dermatologic patients. Front Immunol 2023; 14:1138765. [PMID: 37006291 PMCID: PMC10050596 DOI: 10.3389/fimmu.2023.1138765] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundBy depleting circulating B lymphocytes, rituximab time-dependently suppresses coronavirus disease 2019 (COVID-19) vaccines’ humoral immunogenicity for a prolonged period. The optimal time to vaccinate rituximab-exposed immune-mediated dermatologic disease (IMDD) patients is currently unclear.ObjectiveTo estimate the vaccination timeframe that equalized the occurrence of humoral immunogenicity outcomes between rituximab-exposed and rituximab-naïve IMDD patients.MethodsThis retrospective cohort study recruited rituximab-exposed and age-matched rituximab-naïve subjects tested for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific immunity post-vaccination. Baseline clinical and immunological data (i.e., immunoglobulin levels, lymphocyte immunophenotyping) and SARS-CoV-2-specific immunity levels were extracted. The outcomes compared were the percentages of subjects who produced neutralizing antibodies (seroconversion rates, SR) and SARS-CoV-2-specific IgG levels among seroconverters. The outcomes were first analyzed using multiple regressions adjusted for the effects of corticosteroid use, steroid-spearing agents, and pre-vaccination immunological status (i.e., IgM levels, the percentages of the total, naïve, and memory B lymphocytes) to identify rituximab-related immunogenicity outcomes. The rituximab-related outcome differences with a 95% confidence interval (CI) between groups were calculated, starting by including every subject and then narrowing down to those with longer rituximab-to-vaccination intervals (≥3, ≥6, ≥9, ≥12 months). The desirable cut-off performances were <25% outcome inferiority observed among rituximab-exposed subgroups compared to rituximab-naïve subjects, and the positive likelihood ratio (LR+) for the corresponding outcomes ≥2.FindingsForty-five rituximab-exposed and 90 rituximab-naive subjects were included. The regression analysis demonstrated a negative association between rituximab exposure status and SR but not with SARS-CoV-2-specific IgG levels. Nine-month rituximab-to-vaccination cut-off fulfilled our prespecified diagnostic performance (SR difference between rituximab-exposed and rituximab-naïve group [95%CI]: -2.6 [-23.3, 18.1], LR+: 2.6) and coincided with the repopulation of naïve B lymphocytes in these patients.ConclusionsNine months of rituximab-to-vaccination interval maximize the immunological benefits of COVID-19 vaccines while avoiding unnecessary delay in vaccination and rituximab treatment for IMDD patients.
Collapse
Affiliation(s)
- Chutima Seree-aphinan
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Yanisa Ratanapokasatit
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Poonkiat Suchonwanit
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ploysyne Rattanakaemakorn
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pichaya O-Charoen
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Bangkok, Thailand
| | - Thanitta Suangtamai
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Bangkok, Thailand
| | - Chavachol Setthaudom
- Immunology Laboratory, Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sonphet Chirasuthat
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kumutnart Chanprapaph
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- *Correspondence: Kumutnart Chanprapaph,
| |
Collapse
|
12
|
Sabatino JJ, Mittl K, Rowles W, Zamecnik CR, Loudermilk RP, Gerungan C, Spencer CM, Sagan SA, Alexander J, Mcpolin K, Chen P, Deshpande C, Wyse K, Maiese EM, Wilson MR, Zamvil SS, Bove R. Longitudinal adaptive immune responses following sequential SARS-CoV-2 vaccinations in MS patients on anti-CD20 therapies and sphingosine-1-phosphate receptor modulators. Mult Scler Relat Disord 2023; 70:104484. [PMID: 36608538 PMCID: PMC9794398 DOI: 10.1016/j.msard.2022.104484] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Adequate response to the SARS-CoV-2 vaccine represents an important treatment goal in caring for patients with multiple sclerosis (MS) during the ongoing COVID-19 pandemic. Previous data so far have demonstrated lower spike-specific IgG responses following two SARS-CoV-2 vaccinations in MS patients treated with sphingosine-1-phosphate (S1P) receptor modulators and anti-CD20 monoclonal antibodies (mAb) compared to other disease modifying therapies (DMTs). It is unknown whether subsequent vaccinations can augment antibody responses in these patients. OBJECTIVES The goal of this observational study was to determine the effects of a third SARS-CoV-2 vaccination on antibody and T cell responses in MS patients treated with anti-CD20 mAb or S1P receptor modulators. METHODS Vaccine responses in patients treated with anti-CD20 antibodies (ocrelizumab and ofatumumab) or S1P receptor modulators (fingolimod and siponimod) were evaluated before and after third SARS-CoV-2 vaccination as part of an ongoing longitudinal study. Total spike protein and spike receptor binding domain (RBD)-specific IgG responses were measured by Luminex bead-based assay. Spike-specific CD4+ and CD8+ T cell responses were measured by activation-induced marker expression. RESULTS MS patients and healthy controls were enrolled before and following SARS-CoV-2 vaccination. A total of 31 MS patients (n = 10 ofatumumab, n = 13 ocrelizumab, n = 8 S1P) and 10 healthy controls were evaluated through three SARS-CoV-2 vaccinations. Compared to healthy controls, total spike IgG was significantly lower in anti-CD20 mAb-treated patients and spike RBD IgG was significantly lower in anti-CD20 mAb and S1P-treated patients following a third vaccination. While seropositivity was 100% in healthy controls after a third vaccination, total spike IgG and spike RBD IgG seropositivity were lower in ofatumumab (60% and 60%, respectively), ocrelizumab (85% and 46%, respectively), and S1P-treated patients (100% and 75%, respectively). Longer treatment duration, including prior treatment history, appeared to negatively impact antibody responses. Spike-specific CD4+ and CD8+ T cell responses were well maintained across all groups following a third vaccination. Finally, immune responses were also compared in patients who were vaccinated prior to or following ofatumumab treatment. Antibody responses were significantly higher in those patients who received their primary SARS-CoV-2 vaccination prior to initiating ofatumumab treatment. CONCLUSIONS This study adds to the evolving understanding of SARS-CoV-2 vaccine responses in people with MS treated with disease-modifying therapies (DMTs) known to suppress humoral immunity. Our findings provide important information for optimizing vaccine immunity in at-risk MS patient populations.
Collapse
Affiliation(s)
- Joseph J Sabatino
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kristen Mittl
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - William Rowles
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Colin R Zamecnik
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rita P Loudermilk
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chloe Gerungan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Collin M Spencer
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sharon A Sagan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jessa Alexander
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kira Mcpolin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - PeiXi Chen
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Kerri Wyse
- Novartis Pharmaceuticals, East Hanover, NJ, USA
| | | | - Michael R Wilson
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Scott S Zamvil
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Riley Bove
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
13
|
Titers of antibodies against ancestral SARS-CoV-2 correlate with levels of neutralizing antibodies to multiple variants. NPJ Vaccines 2022; 7:174. [PMID: 36585405 PMCID: PMC9801350 DOI: 10.1038/s41541-022-00586-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022] Open
Abstract
Diagnostic assays currently used to monitor the efficacy of COVID-19 vaccines measure levels of antibodies to the receptor-binding domain of ancestral SARS-CoV-2 (RBDwt). However, the predictive value for protection against new variants of concern (VOCs) has not been firmly established. Here, we used bead-based arrays and flow cytometry to measure binding of antibodies to spike proteins and receptor-binding domains (RBDs) from VOCs in 12,000 serum samples. Effects of sera on RBD-ACE2 interactions were measured as a proxy for neutralizing antibodies. The samples were obtained from healthy individuals or patients on immunosuppressive therapy who had received two to four doses of COVID-19 vaccines and from COVID-19 convalescents. The results show that anti-RBDwt titers correlate with the levels of binding- and neutralizing antibodies against the Alpha, Beta, Gamma, Delta, Epsilon and Omicron variants. The benefit of multiplexed analysis lies in the ability to measure a wide range of anti-RBD titers using a single dilution of serum for each assay. The reactivity patterns also yield an internal reference for neutralizing activity and binding antibody units per milliliter (BAU/ml). Results obtained with sera from vaccinated healthy individuals and patients confirmed and extended results from previous studies on time-dependent waning of antibody levels and effects of immunosuppressive agents. We conclude that anti-RBDwt titers correlate with levels of neutralizing antibodies against VOCs and propose that our method may be implemented to enhance the precision and throughput of immunomonitoring.
Collapse
|
14
|
Bazzi SA, Maguire C, Holay N, Geltman J, Hurley K, DiPasquale C, Abigania M, Olson E, Ehrlich LIR, Triplett TA, Melamed E. Longitudinal COVID-19 immune trajectories in patients with neurological autoimmunity on anti-CD20 therapy. Mult Scler Relat Disord 2022; 68:104195. [PMID: 36223705 PMCID: PMC9511881 DOI: 10.1016/j.msard.2022.104195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES During the COVID-19 pandemic, B cell depleting therapies pose a clinical concern for patients with neuroimmune conditions, as patients may not mount a sufficient immune response to SARS-CoV-2 infection and vaccinations. Studies to-date have reported conflicting results on the degree of antibody production post-SARS-CoV-2 infection and vaccinations in B cell depleted patients, focusing primarily on short-term immune profiling. Our objective was to follow longitudinal immune responses in COVID-19 B cell depleted patients with neuroimmune disorders post-COVID-19 and SARS-CoV-2-vaccination. METHODS CD20 B cell depleted autoimmune patients and age/sex-matched controls positive for SARS-CoV-2 were recruited at Dell Medical School, UT Austin between 2020 and 2021, followed prospectively for 12 months and evaluated at multiple time points for spike S1 receptor binding domain (RBD) antibody titers, B and T cell composition, and frequency of T cells specific for SARS-CoV-2 antigens. RESULTS Immune responses post-SARS-CoV-2 infection and vaccination were evaluated in a cohort of COVID-19 B cell depleted neuroimmune patients (n = 5), COVID-19 non-B cell depleted autoimmune patients (n = 15), COVID-19 immunocompetent patients (n = 117), and healthy controls (n = 6) for a total of 259 samples in 137 participants. 4/5 B cell-depleted patients developed detectable anti-spike RBD antibodies, which were boosted by vaccination in 2 patients. While spike RBD antibodies were associated with presence of CD20+ B cells, very few B cells were required. In contrast, patients whose B cell compartment primarily consisted of CD19+CD20- Bcells during acute COVID-19 disease or vaccination did not seroconvert. Interestingly, circulating Bcells in B cell depleted patients were significantly CD38high with co-expression of CD24 and CD27, indicating that B cell depletion may impact B cell activation patterns. Additionally, all B cell depleted patients mounted a sustained T cell response to SARS-CoV-2 antigens, regardless of seroconversion. Specifically, all patients developed naïve, central memory, effector memory, and effector memory RA+ T cells, suggesting intact T cell memory conversion in B cell depleted patients compared to controls. DISCUSSION We present the longest COVID-19 immune profiling analysis to date in B cell depleted patients, demonstrating that both humoral and cellular immune responses can be generated and sustained up to 12 months post SARS-CoV-2 infection and vaccination. Notably, failure to establish humoral immunity did not result in severe disease. We also highlight specific T and B cell signatures that could be used as clinical biomarkers to advise patients on timing of SARS-CoV-2 vaccinations.
Collapse
Affiliation(s)
- Sam A Bazzi
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Cole Maguire
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Nisha Holay
- Department of Oncology Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Janelle Geltman
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Kerin Hurley
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | | | | | - Eric Olson
- Babson Diagnostics, Austin, TX, United States
| | - Lauren I R Ehrlich
- Department of Oncology Dell Medical School, University of Texas at Austin, Austin, TX, United States; Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Todd A Triplett
- Department of Oncology Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Esther Melamed
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
15
|
Kister I, Curtin R, Pei J, Perdomo K, Bacon TE, Voloshyna I, Kim J, Tardio E, Velmurugu Y, Nyovanie S, Valeria Calderon A, Dibba F, Stanzin I, Samanovic MI, Raut P, Raposo C, Priest J, Cabatingan M, Winger RC, Mulligan MJ, Patskovsky Y, Silverman GJ, Krogsgaard M. Hybrid and vaccine-induced immunity against SAR-CoV-2 in MS patients on different disease-modifying therapies. Ann Clin Transl Neurol 2022; 9:1643-1659. [PMID: 36165097 PMCID: PMC9538694 DOI: 10.1002/acn3.51664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To compare "hybrid immunity" (prior COVID-19 infection plus vaccination) and post-vaccination immunity to SARS CoV-2 in MS patients on different disease-modifying therapies (DMTs) and to assess the impact of vaccine product and race/ethnicity on post-vaccination immune responses. METHODS Consecutive MS patients from NYU MS Care Center (New York, NY), aged 18-60, who completed primary COVID-19 vaccination series ≥6 weeks previously were evaluated for SARS CoV-2-specific antibody responses with electro-chemiluminescence and multiepitope bead-based immunoassays and, in a subset, live virus immunofluorescence-based microneutralization assay. SARS CoV-2-specific cellular responses were assessed with cellular stimulation TruCulture IFNγ and IL-2 assay and, in a subset, with IFNγ and IL-2 ELISpot assays. Multivariate analyses examined associations between immunologic responses and prior COVID-19 infection while controlling for age, sex, DMT at vaccination, time-to-vaccine, and vaccine product. RESULTS Between 6/01/2021 and 11/11/2021, 370 MS patients were recruited (mean age 40.6 years; 76% female; 53% non-White; 22% with prior infection; common DMT classes: ocrelizumab 40%; natalizumab 15%, sphingosine-1-phosphate receptor modulators 13%; and no DMT 8%). Vaccine-to-collection time was 18.7 (±7.7) weeks and 95% of patients received mRNA vaccines. In multivariate analyses, patients with laboratory-confirmed prior COVID-19 infection had significantly increased antibody and cellular post-vaccination responses compared to those without prior infection. Vaccine product and DMT class were independent predictors of antibody and cellular responses, while race/ethnicity was not. INTERPRETATION Prior COVID-19 infection is associated with enhanced antibody and cellular post-vaccine responses independent of DMT class and vaccine type. There were no differences in immune responses across race/ethnic groups.
Collapse
Affiliation(s)
- Ilya Kister
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Ryan Curtin
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Jinglan Pei
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | - Katherine Perdomo
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Tamar E. Bacon
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Iryna Voloshyna
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Joseph Kim
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Ethan Tardio
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Yogambigai Velmurugu
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Samantha Nyovanie
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Andrea Valeria Calderon
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Fatoumatta Dibba
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Igda Stanzin
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Marie I. Samanovic
- NYU Langone Vaccine Center, Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Pranil Raut
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | | | | | | | - Mark J. Mulligan
- NYU Langone Vaccine Center, Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Yury Patskovsky
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Gregg J. Silverman
- Division of Rheumatology, Department of MedicineNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| |
Collapse
|
16
|
Meca-Lallana V, García Domínguez JM, López Ruiz R, Martín-Martínez J, Arés Luque A, Hernández Pérez MA, Prieto González JM, Landete Pascual L, Sastre-Garriga J. Expert-Agreed Practical Recommendations on the Use of Cladribine. Neurol Ther 2022; 11:1475-1488. [PMID: 36068429 PMCID: PMC9447968 DOI: 10.1007/s40120-022-00394-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/03/2022] [Indexed: 12/13/2022] Open
Abstract
Cladribine is a disease-modifying selective immune reconstitution oral therapy for adult patients with highly active relapsing multiple sclerosis (RMS). It was approved in the USA in 2019 and in Europe in 2017, thus there are still gaps in existing guidelines for using cladribine tablets in clinical practice. Nine experts with extensive experience in managing patients with multiple sclerosis in Spain identified some of the unanswered questions related to the real-life use of cladribine tablets. They reviewed the available clinical trial data and real-world evidence, including their own experiences of using cladribine, over the course of three virtual meetings held between November 2020 and January 2021. This article gathers their practical recommendations to aid treatment decision-making and optimise the use of cladribine tablets in patients with RMS. The consensus recommendations cover the following areas: candidate patient profiles, switching strategies (to and from cladribine), managing response to cladribine and safety considerations.
Collapse
Affiliation(s)
- Virginia Meca-Lallana
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario "La Princesa", Madrid, Spain.
| | | | - Rocío López Ruiz
- Multiple Sclerosis Unit, Hospital Universitario Virgen Macarena, Seville, Spain
| | | | - Adrián Arés Luque
- Neurology Department, Complejo Asistencial Universitario de León, León, Spain
| | | | | | | | - Jaume Sastre-Garriga
- Neurology Department, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
17
|
Etemadifar M, Nouri H, Pitzalis M, Idda ML, Salari M, Baratian M, Mahdavi S, Abhari AP, Sedaghat N. Multiple sclerosis disease-modifying therapies and COVID-19 vaccines: a practical review and meta-analysis. J Neurol Neurosurg Psychiatry 2022; 93:986-994. [PMID: 35688629 DOI: 10.1136/jnnp-2022-329123] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
Abstract
Studies among people with multiple sclerosis (pwMS) receiving disease-modifying therapies (DMTs) have provided adequate evidence for an appraisal of COVID-19 vaccination policies among them. To synthesise the available evidence addressing the effect of MS DMTs on COVID-19 vaccines' immunogenicity and effectiveness, following the Cochrane guidelines, we systematically reviewed all observational studies available in MEDLINE, Scopus, Web of Science, MedRxiv and Google Scholar from January 2021 to January 2022 and extracted their relevant data. Immunogenicity data were then synthesised in a quantitative, and other data in a qualitative manner. Evidence from 28 studies suggests extensively lower B-cell responses in sphingosine-1-phosphate receptor modulator (S1PRM) treated and anti-CD20 (aCD20) treated, and lower T-cell responses in interferon-treated, S1PRM-treated and cladribine-treated pwMS-although most T cell evidence currently comprises of low or very low certainty. With every 10-week increase in aCD20-to-vaccine period, a 1.94-fold (95% CI 1.57 to 2.41, p<0.00001) increase in the odds of seroconversion was observed. Furthermore, the evidence points out that B-cell-depleting therapies may accelerate postvaccination humoral waning, and boosters' immunogenicity is predictable with the same factors affecting the initial vaccination cycle. Four real-world studies further indicate that the comparative incidence/severity of breakthrough COVID-19 has been higher among the pwMS treated with S1PRM and aCD20-unlike the ones treated with other DMTs. S1PRM and aCD20 therapies were the only DMTs reducing the real-world effectiveness of COVID-19 vaccination among pwMS. Hence, it could be concluded that optimisation of humoral immunogenicity and ensuring its durability are the necessities of an effective COVID-19 vaccination policy among pwMS who receive DMTs.
Collapse
Affiliation(s)
- Masoud Etemadifar
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hosein Nouri
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Maristella Pitzalis
- Institute of Genetic and Biomedical Research (IRGB) of the National Research Council (CNR), Cagliari, Italy
| | - Maria Laura Idda
- Institute of Genetic and Biomedical Research (IRGB) of the National Research Council (CNR), Cagliari, Italy
| | - Mehri Salari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Baratian
- Clinical Research Developement Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Sepide Mahdavi
- Clinical Research Developement Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Amir Parsa Abhari
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Nahad Sedaghat
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran .,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| |
Collapse
|
18
|
Mrak D, Simader E, Sieghart D, Mandl P, Radner H, Perkmann T, Haslacher H, Mayer M, Koblischke M, Hofer P, Göschl L, Kartnig F, Deimel T, Kerschbaumer A, Hummel T, Kornek B, Thalhammer R, Stiasny K, Winkler S, Smolen JS, Aberle JH, Aletaha D, Heinz LX, Bonelli M. Immunogenicity and safety of a fourth COVID-19 vaccination in rituximab-treated patients: an open-label extension study. Ann Rheum Dis 2022; 81:1750-1756. [PMID: 35977809 DOI: 10.1136/ard-2022-222579] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Patients under rituximab therapy are at high risk for a severe COVID-19 disease course. Humoral immune responses to SARS-CoV-2 vaccination are vastly diminished in B-cell-depleted patients, even after a third vaccine dose. However, it remains unclear whether these patients benefit from a fourth vaccination and whether continued rituximab therapy affects antibody development. METHODS In this open-label extension trial, 37 rituximab-treated patients who received a third dose with either a vector or mRNA-based vaccine were vaccinated a fourth time with an mRNA-based vaccine (mRNA-1273 or BNT162b2). Key endpoints included the humoral and cellular immune response as well as safety after a fourth vaccination. RESULTS The number of patients who seroconverted increased from 12/36 (33%) to 21/36 (58%) following the fourth COVID-19 vaccination. In patients with detectable antibodies to the spike protein's receptor-binding domain (median: 8.0 binding antibody units (BAU)/mL (quartiles: 0.4; 13.8)), elevated levels were observed after the fourth vaccination (134.0 BAU/mL (quartiles: 25.5; 1026.0)). Seroconversion and antibody increase were strongly diminished in patients who received rituximab treatment between the third and the fourth vaccination. The cellular immune response declined 12 weeks after the third vaccination, but could only be slightly enhanced by a fourth vaccination. No unexpected safety signals were detected, one serious adverse event not related to vaccination occurred. CONCLUSIONS A fourth vaccine dose is immunogenic in a fraction of rituximab-treated patients. Continuation of rituximab treatment reduced humoral immune response, suggesting that rituximab affects a second booster vaccination. It might therefore be considered to postpone rituximab treatment in clinically stable patients. TRIAL REGISTRATION NUMBER 2021-002348-57.
Collapse
Affiliation(s)
- Daniel Mrak
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Simader
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Peter Mandl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Helga Radner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Margareta Mayer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Philipp Hofer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Lisa Göschl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Felix Kartnig
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Deimel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Hummel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.,2nd Department of Medicine, Lower Austrian Competence Center for Rheumatology, Landesklinikum Stockerau, Stockerau, Lower Austria, Austria
| | - Barbara Kornek
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Stefan Winkler
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Judith H Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Cauchi M, Willis M, Andrews A, Backx M, Brownlee W, Ford HL, Gran B, Jolles S, Price S, Rashid W, Schmierer K, Tallantyre EC. Multiple sclerosis and the risk of infection: Association of British Neurologists consensus guideline. Pract Neurol 2022; 22:practneurol-2022-003370. [PMID: 35863879 DOI: 10.1136/practneurol-2022-003370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
Infection in people with multiple sclerosis (MS) is of major concern, particularly for those receiving disease-modifying therapies. This article explores the risk of infection in people with MS and provides guidance-developed by Delphi consensus by specialists involved in their management-on how to screen for, prevent and manage infection in this population.
Collapse
Affiliation(s)
- Marija Cauchi
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, University Hospital of Wales, Cardiff, UK
| | - Mark Willis
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, University Hospital of Wales, Cardiff, UK
| | - Angela Andrews
- Pharmacy Neurosciences Directorate, University Hospital of Wales, Cardiff, UK
| | - Matthijs Backx
- Infectious Diseases, University Hospital of Wales and Department of Microbiology, Public Health Wales, Cardiff, UK
| | - Wallace Brownlee
- Queen Square MS Centre, University College London Institute of Neurology, Queen Square Multiple Sclerosis Centre, London, UK
| | - Helen L Ford
- Centre for Neurosciences, Leeds Teaching Hospitals NHS Trust, Leeds, UK, Leeds, UK
| | - Bruno Gran
- Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham, UK
- Mental Health and Clinical Neuroscience Academic Unit, University of Nottingham School of Medicine, Nottingham, UK
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Sian Price
- Department of Neuroscience, University of Sheffield, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Waqar Rashid
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Klaus Schmierer
- The Blizard Institute (Neuroscience, Surgery & Trauma), Queen Mary University of London Faculty of Medicine and Dentistry, London, UK
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Emma C Tallantyre
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
20
|
Abstract
Purpose of Review COVID-19 has posed a continuously evolving challenge for providers caring for patients with multiple sclerosis (MS). While guidelines from national and international organizations came quickly, these have required constant reassessment and modification as the pandemic has progressed. This review aims to assess the first 2 years of literature on COVID-19 relevant to the clinical management of patients with MS. In particular, we will review how MS impacts the risk of COVID-19 infection, how disease-modifying therapies may alter this risk, and explore considerations regarding disease-modifying therapy (DMT) and vaccination for COVID-19. We will also explore potential ways in which a COVID-19 infection may impact multiple sclerosis. Our goal is to provide an overarching review of the major findings at this stage of the pandemic relevant to those that care for patients with MS. Recent Findings Over the course of the COVID-19 pandemic, providers have had to re-evaluate the priorities in the management of MS. A growing number of studies have evaluated the relevant risk factors and considerations regarding MS and particular disease-modifying therapies. Summary The long-term impacts of the pandemic on the health of those with MS will continue to be revealed. In general, most patients with MS do not need major revisions to their treatment plan due to COVID-19 risk. However, individuals who are older, more disabled, and on more potent therapies may need to consider strategies for decreasing their overall risk. Regardless, continued improvement in our understanding of interactions between infections, disease-modifying therapy, and MS are paramount to optimizing the care of those with MS going forward.
Collapse
|
21
|
Baker D, MacDougall A, Kang AS, Schmierer K, Giovannoni G, Dobson R. Seroconversion following COVID-19 vaccination: can we optimize protective response in CD20-treated individuals? Clin Exp Immunol 2022; 207:263-271. [PMID: 35553629 PMCID: PMC9113152 DOI: 10.1093/cei/uxab015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Although there is an ever-increasing number of disease-modifying treatments for relapsing multiple sclerosis (MS), few appear to influence coronavirus disease 2019 (COVID-19) severity. There is concern about the use of anti-CD20-depleting monoclonal antibodies, due to the apparent increased risk of severe disease following severe acute respiratory syndrome corona virus two (SARS-CoV-2) infection and inhibition of protective anti-COVID-19 vaccine responses. These antibodies are given as maintenance infusions/injections and cause persistent depletion of CD20+ B cells, notably memory B-cell populations that may be instrumental in the control of relapsing MS. However, they also continuously deplete immature and mature/naïve B cells that form the precursors for infection-protective antibody responses, thus blunting vaccine responses. Seroconversion and maintained SARS-CoV-2 neutralizing antibody levels provide protection from COVID-19. However, it is evident that poor seroconversion occurs in the majority of individuals following initial and booster COVID-19 vaccinations, based on standard 6 monthly dosing intervals. Seroconversion may be optimized in the anti-CD20-treated population by vaccinating prior to treatment onset or using extended/delayed interval dosing (3-6 month extension to dosing interval) in those established on therapy, with B-cell monitoring until (1-3%) B-cell repopulation occurs prior to vaccination. Some people will take more than a year to replete and therefore protection may depend on either the vaccine-induced T-cell responses that typically occur or may require prophylactic, or rapid post-infection therapeutic, antibody or small-molecule antiviral treatment to optimize protection against COVID-19. Further studies are warranted to demonstrate the safety and efficacy of such approaches and whether or not immunity wanes prematurely as has been observed in the other populations.
Collapse
Affiliation(s)
- David Baker
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Amy MacDougall
- Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Angray S Kang
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Centre for Oral Immunobiology and Regenerative Medicine, Dental Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Klaus Schmierer
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Gavin Giovannoni
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Ruth Dobson
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, Barts and The London School of Medicine & Dentistry, London, UK
| |
Collapse
|
22
|
Syversen SW, Jyssum I, Tveter AT, Tran TT, Sexton J, Provan SA, Mjaaland S, Warren DJ, Kvien TK, Grødeland G, Nissen‐Meyer LSH, Ricanek P, Chopra A, Andersson AM, Kro GB, Jahnsen J, Munthe LA, Haavardsholm EA, Vaage JT, Lund‐Johansen F, Jørgensen KK, Goll GL. Immunogenicity and Safety of Standard and Third Dose SARS-CoV-2 Vaccination in Patients on Immunosuppressive Therapy. Arthritis Rheumatol 2022; 74:1321-1332. [PMID: 35507355 PMCID: PMC9347774 DOI: 10.1002/art.42153] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/29/2022] [Accepted: 04/28/2022] [Indexed: 11/10/2022]
Abstract
Objective Immunogenicity and safety following receipt of the standard SARS–CoV‐2 vaccination regimen in patients with immune‐mediated inflammatory diseases (IMIDs) are poorly characterized, and data after receipt of the third vaccine dose are lacking. The aim of the study was to evaluate serologic responses and adverse events following the standard 2‐dose regimen and a third dose of SARS–CoV‐2 vaccine in IMID patients receiving immunosuppressive therapy. Methods Adult patients receiving immunosuppressive therapy for rheumatoid arthritis, spondyloarthritis, psoriatic arthritis, Crohn's disease, or ulcerative colitis, as well as healthy adult controls, who received the standard 2‐dose SARS–CoV‐2 vaccination regimen were included in this prospective observational study. Analyses of antibodies to the receptor‐binding domain (RBD) of the SARS–CoV‐2 spike protein were performed prior to and 2–4 weeks after vaccination. Patients with a weak serologic response, defined as an IgG antibody titer of ≤100 arbitrary units per milliliter (AU/ml) against the receptor‐binding domain of the full‐length SARS–Cov‐2 spike protein, were allotted a third vaccine dose. Results A total of 1,505 patients (91%) and 1,096 healthy controls (98%) had a serologic response to the standard regimen (P < 0.001). Anti‐RBD antibody levels were lower in patients (median 619 AU/ml interquartile range [IQR] 192–4,191) than in controls (median 3,355 AU/ml [IQR 896–7,849]) (P < 0.001). The proportion of responders was lowest among patients receiving tumor necrosis factor inhibitor combination therapy, JAK inhibitors, or abatacept. Younger age and receipt of messenger RNA–1273 vaccine were predictors of serologic response. Of 153 patients who had a weak response to the standard regimen and received a third dose, 129 (84%) became responders. The vaccine safety profile among patients and controls was comparable. Conclusion IMID patients had an attenuated response to the standard vaccination regimen as compared to healthy controls. A third vaccine dose was safe and resulted in serologic response in most patients. These data facilitate identification of patient groups at risk of an attenuated vaccine response, and they support administering a third vaccine dose to IMID patients with a weak serologic response to the standard regimen.
Collapse
Affiliation(s)
| | - Ingrid Jyssum
- Diakonhjemmet Hospital and University of OsloOsloNorway
| | | | | | | | | | | | | | - Tore K. Kvien
- Diakonhjemmet Hospital and University of OsloOsloNorway
| | | | | | | | | | | | | | - Jørgen Jahnsen
- University of Oslo, Oslo, and Akershus University HospitalLørenskogNorway
| | | | | | - John T. Vaage
- Oslo University Hospital and University of OsloOsloNorway
| | | | | | | |
Collapse
|
23
|
Tolf A, Wiberg A, Müller M, Nazir FH, Pavlovic I, Laurén I, Mangsbo S, Burman J. Factors Associated With Serological Response to SARS-CoV-2 Vaccination in Patients With Multiple Sclerosis Treated With Rituximab. JAMA Netw Open 2022; 5:e2211497. [PMID: 35544139 PMCID: PMC9096596 DOI: 10.1001/jamanetworkopen.2022.11497] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/24/2022] [Indexed: 12/14/2022] Open
Abstract
Importance B-cell-depleting monoclonal antibodies are widely used for treatment of multiple sclerosis but are associated with an impaired response to vaccines. Objective To identify factors associated with a favorable vaccine response to tozinameran. Design, Setting, and Participants This prospective cohort study was conducted in a specialized multiple sclerosis clinic at a university hospital from January 21 to December 1, 2021. Of 75 patients evaluated for participation who received a diagnosis of multiple sclerosis with planned or ongoing treatment with rituximab, 69 were included in the study, and data from 67 were analyzed. Exposures Sex, age, number of previous rituximab infusions, accumulated dose of rituximab, previous COVID-19 infection, time since last rituximab treatment, CD19+ B-cell count before vaccination, CD4+ T-cell count, and CD8+ T-cell count were considered potential factors associated with the main outcome. Main Outcomes and Measures Serological vaccine responses were measured by quantitation of anti-spike immunoglobulin G (IgG) antibodies, anti-receptor-binding domain (RBD) IgG antibodies, and their neutralizing capacities. Cellular responses to spike protein-derived SARS-CoV-2 peptide pools were assessed by counting interferon gamma spot-forming units in a FluoroSpot assay. Results Among 60 patients with ongoing rituximab treatment (49 women [82%]; mean (SD) age, 43 [10] years), the median (range) disease duration was 9 (1-29) years, and the median (range) dose of rituximab was 2750 (500-10 000) mg during a median (range) time of 2.8 (0.5-8.3) years. The median (range) follow-up from the first vaccination dose was 7.3 (4.3-10.0) months. Vaccine responses were determined before vaccination with tozinameran and 6 weeks after vaccination. By using established cutoff values for anti-spike IgG (264 binding antibody units/mL) and anti-RBD IgG (506 binding antibody units/mL), the proportion of patients with a positive response increased with the number of B cells, which was the only factor associated with these outcomes. A cutoff for the B-cell count of at least 40/μL was associated with an optimal serological response. At this cutoff, 26 of 29 patients (90%) had positive test results for anti-spike IgG and 21 of 29 patients (72%) for anti-RBD IgG, and 27 of 29 patients (93%) developed antibodies with greater than 90% inhibition of angiotensin-converting enzyme 2. No factor associated with the cellular response was identified. Depending on the peptide pool, 21 of 25 patients (84%) to 22 of 25 patients (88%) developed a T-cell response with interferon gamma production at the B-cell count cutoff of at least 40/μL. Conclusions and Relevance This cohort study found that for an optimal vaccine response from tozinameran, rituximab-treated patients with multiple sclerosis may be vaccinated as soon as possible, with rituximab treatment delayed until B-cell counts have reached at least 40/μL. An additional vaccination with tozinameran should be considered at that point.
Collapse
Affiliation(s)
- Andreas Tolf
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Anna Wiberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Malin Müller
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Ivan Pavlovic
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ida Laurén
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Abstract
Neuroimmunological diseases and their treatment compromise the immune system, thereby increasing the risk of infections and serious illness. Consequently, vaccinations to protect against infections are an important part of the clinical management of these diseases. However, the wide variety of immunotherapies that are currently used to treat neuroimmunological disease — particularly multiple sclerosis and neuromyelitis optica spectrum disorders — can also impair immunological responses to vaccinations. In this Review, we discuss what is known about the effects of various immunotherapies on immunological responses to vaccines and what these effects mean for the safe and effective use of vaccines in patients with a neuroimmunological disease. The success of vaccination in patients receiving immunotherapy largely depends on the specific mode of action of the immunotherapy. To minimize the risk of infection when using immunotherapy, assessment of immune status and exclusion of underlying chronic infections before initiation of therapy are essential. Selection of the required vaccinations and leaving appropriate time intervals between vaccination and administration of immunotherapy can help to safeguard patients. We also discuss the rapidly evolving knowledge of how immunotherapies affect responses to SARS-CoV-2 vaccines and how these effects should influence the management of patients on these therapies during the COVID-19 pandemic. In this Review, the authors discuss how various immunotherapies for neuroimmunological diseases interact with vaccination responses, including responses to SARS-CoV-2 vaccinations, and the implications for the safe and effective use of vaccines in patients with these diseases. Vaccination against infection is an essential part of the management of neuroimmunological diseases. All indicated vaccinations should be administered before initiation of immunotherapy whenever possible; appropriate intervals between vaccination and treatment vary with treatment and vaccination. Inactivated vaccines are considered safe in neuroimmunological diseases but live vaccines are generally contraindicated during immunotherapy. Vaccination responses during immunotherapy can be diminished or abrogated, depending on the treatment and vaccination; antibody titre testing to monitor responses can be considered where appropriate. Vaccinations must be avoided during relapses or exacerbations of neuroimmunological diseases. Vaccination against SARS-CoV-2 is recommended for patients with neuroimmunological disease but some immunotherapies limit the immune response; therefore, timing should be considered carefully.
Collapse
|
25
|
Töllner M, Speer C, Benning L, Bartenschlager M, Nusshag C, Morath C, Zeier M, Süsal C, Schnitzler P, Schmitt W, Bergner R, Bartenschlager R, Lorenz HM, Schaier M. Impaired Neutralizing Antibody Activity against B.1.617.2 (Delta) after Anti-SARS-CoV-2 Vaccination in Patients Receiving Anti-CD20 Therapy. J Clin Med 2022; 11:jcm11061739. [PMID: 35330069 PMCID: PMC8952324 DOI: 10.3390/jcm11061739] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022] Open
Abstract
Background: To characterize humoral response after standard anti-SARS-CoV-2 vaccination in Rituximab-treated patients and to determine the optimal time point after last Rituximab treatment for appropriate immunization. Methods: Sixty-four patients who received Rituximab within the last seven years prior to the first anti-SARS-CoV-2 vaccination were recruited in a prospective observational study. Anti-S1 IgG, SARS-CoV-2 specific neutralization, and various SARS-CoV-2 target antibodies were determined. A live virus assay was used to assess neutralizing antibody activity against B.1.617.2 (delta). In Rituximab-treated patients, CD19+ peripheral B-cells were quantified using flow cytometry. Results: After second vaccination, all antibodies were significantly reduced compared to healthy controls. Neutralizing antibody activity against B.1.617.2 (delta) was detectable with a median (IQR) ID50 of 0 (0−1:20) compared to 1:320 (1:160−1:320) in healthy controls (for all p < 0.001). Longer time period since last Rituximab administration correlated with higher anti-SARS-CoV-2 antibody levels and a stronger neutralization of B.1.617.2 (delta). With one exception, only patients with a CD19+ cell proportion ≥ 1% had detectable neutralizing antibodies. Conclusion: Our data indicate that a reconstitution of the B-cell population to >1% seems crucial in developing neutralizing antibodies against SARS-CoV-2. We suggest that anti-SARS-CoV-2 vaccination should be administered at least 8−12 months after the last Rituximab treatment for sufficient humoral responses.
Collapse
Affiliation(s)
- Maximilian Töllner
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Correspondence:
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Molecular Medicine Partnership Unit Heidelberg, European Molecular Biology Laboratory, 69120 Heidelberg, Germany
| | - Louise Benning
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Christian Morath
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Caner Süsal
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul 34010, Turkey;
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University of Heidelberg, 69120 Heidelberg, Germany;
| | | | - Raoul Bergner
- Clinical Center Ludwigshafen, Department of Internal Medicine A, 67036 Ludwigshafen, Germany;
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
- German Center for Infection Research (DZIF), Heidelberg Partner Site, 69120 Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| |
Collapse
|
26
|
Kister I, Patskovsky Y, Curtin R, Pei J, Perdomo K, Rimler Z, Voloshyna I, Samanovic MI, Cornelius AR, Velmurugu Y, Nyovanie S, Kim J, Tardio E, Bacon TE, Zhovtis Ryerson L, Raut P, Rosetta P, Hawker K, Raposo C, Priest J, Cabatingan M, Winger RC, Mulligan MJ, Krogsgaard M, Silverman GJ. Cellular and humoral immunity to SARS-CoV-2 infection in multiple sclerosis patients on ocrelizumab and other disease-modifying therapies: a multi-ethnic observational study. Ann Neurol 2022; 91:782-795. [PMID: 35289960 PMCID: PMC9082484 DOI: 10.1002/ana.26346] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To determine the impact of MS disease-modifying therapies (DMTs) on the development of cellular and humoral immunity to SARS-CoV-2 infection. METHODS MS patients aged 18-60 were evaluated for anti-nucleocapsid and anti-Spike RBD antibody with electro-chemiluminescence immunoassay; antibody responses to Spike protein, RBD, N-terminal domain with multiepitope bead-based immunoassays (MBI); live virus immunofluorescence-based microneutralization assay; T-cell responses to SARS-CoV-2 Spike using TruCulture ELISA; and IL-2 and IFNγ ELISpot assays. Assay results were compared by DMT class. Spearman correlation and multivariate analyses were performed to examine associations between immunologic responses and infection severity. RESULTS Between 1/6/2021 and 7/21/2021, 389 MS patients were recruited (mean age 40.3 years; 74% female; 62% non-White). Most common DMTs were ocrelizumab (OCR) - 40%; natalizumab - 17%, Sphingosine 1-phosphate receptor (S1P) modulators -12%; and 15% untreated. 177 patients (46%) had laboratory evidence of SARS-CoV-2 infection; 130 had symptomatic infection, 47 - asymptomatic. Antibody responses were markedly attenuated in OCR compared to other groups (p≤0.0001). T-cell responses (IFNγ) were decreased in S1P (p=0.03), increased in natalizumab (p<0.001), and similar in other DMTs, including OCR. Cellular and humoral responses were moderately correlated in both OCR (r=0.45, p=0.0002) and non-OCR (r=0.64, p<0.0001). Immune responses did not differ by race/ethnicity. COVID-19 clinical course was mostly non-severe and similar across DMTs; 7% (9/130) were hospitalized. INTERPRETATION DMTs had differential effects on humoral and cellular immune responses to SARS-CoV-2 infection. Immune responses did not correlate with COVID-19 clinical severity in this relatively young and non-disabled group of MS patients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ilya Kister
- NYU Multiple Sclerosis Comprehensive Care Center, Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Yury Patskovsky
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ryan Curtin
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jinglan Pei
- Genentech, Inc., South San Francisco, CA, USA
| | - Katherine Perdomo
- NYU Multiple Sclerosis Comprehensive Care Center, Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Zoe Rimler
- NYU Multiple Sclerosis Comprehensive Care Center, Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Iryna Voloshyna
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Marie I Samanovic
- NYU Langone Vaccine Center, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Amber R Cornelius
- NYU Langone Vaccine Center, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Yogambigai Velmurugu
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Samantha Nyovanie
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Joseph Kim
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ethan Tardio
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Tamar E Bacon
- NYU Multiple Sclerosis Comprehensive Care Center, Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Lana Zhovtis Ryerson
- NYU Multiple Sclerosis Comprehensive Care Center, Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Pranil Raut
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | - Mark J Mulligan
- NYU Langone Vaccine Center, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Gregg J Silverman
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
27
|
König M, Torgauten HM, Tran TT, Holmøy T, Vaage JT, Lund-Johansen F, Nygaard GO. Immunogenicity and Safety of a Third SARS-CoV-2 Vaccine Dose in Patients With Multiple Sclerosis and Weak Immune Response After COVID-19 Vaccination. JAMA Neurol 2022; 79:307-309. [PMID: 35072702 PMCID: PMC8787678 DOI: 10.1001/jamaneurol.2021.5109] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/29/2021] [Indexed: 07/23/2023]
Abstract
This cohort study investigates the immunogenicity and safety of a third SARS-CoV-2 vaccine dose in patients with multiple sclerosis who had a weak immune response to COVID-19 vaccination.
Collapse
Affiliation(s)
- Marton König
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Hilde Marie Torgauten
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - The Trung Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
28
|
Jyssum I, Kared H, Tran TT, Tveter AT, Provan SA, Sexton J, Jørgensen KK, Jahnsen J, Kro GB, Warren DJ, Vaage EB, Kvien TK, Nissen-Meyer LSH, Anderson AM, Grødeland G, Haavardsholm EA, Vaage JT, Mjaaland S, Syversen SW, Lund-Johansen F, Munthe LA, Goll GL. Humoral and cellular immune responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis: a prospective, cohort study. THE LANCET. RHEUMATOLOGY 2022; 4:e177-e187. [PMID: 34977602 PMCID: PMC8700278 DOI: 10.1016/s2665-9913(21)00394-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND In rituximab-treated patients with rheumatoid arthritis, humoral and cellular immune responses after two or three doses of SARS-CoV-2 vaccines are not well characterised. We aimed to address this knowledge gap. METHODS This prospective, cohort study (Nor-vaC) was done at two hospitals in Norway. For this sub-study, we enrolled patients with rheumatoid arthritis on rituximab treatment and healthy controls who received SARS-CoV-2 vaccines according to the Norwegian national vaccination programme. Patients with insufficient serological responses to two doses (antibody to the receptor-binding domain [RBD] of the SARS-CoV-2 spike protein concentration <100 arbitrary units [AU]/mL) were allotted a third vaccine dose. Antibodies to the RBD of the SARS-CoV-2 spike protein were measured in serum 2-4 weeks after the second and third doses. Vaccine-elicited T-cell responses were assessed in vitro using blood samples taken before and 7-10 days after the second dose and 3 weeks after the third dose from a subset of patients by stimulating cryopreserved peripheral blood mononuclear cells with spike protein peptides. The main outcomes were the proportions of participants with serological responses (anti-RBD antibody concentrations of ≥70 AU/mL) and T-cell responses to spike peptides following two and three doses of SARS-CoV-2 vaccines. The study is registered at ClinicalTrials.gov, NCT04798625, and is ongoing. FINDINGS Between Feb 9, 2021, and May 27, 2021, 90 patients were enrolled, 87 of whom donated serum and were included in our analyses (69 [79·3%] women and 18 [20·7%] men). 1114 healthy controls were included (854 [76·7%] women and 260 [23·3%] men). 49 patients were allotted a third vaccine dose. 19 (21·8%) of 87 patients, compared with 1096 (98·4%) of 1114 healthy controls, had a serological response after two doses (p<0·0001). Time since last rituximab infusion (median 267 days [IQR 222-324] in responders vs 107 days [80-152] in non-responders) and vaccine type (mRNA-1273 vs BNT162b2) were significantly associated with serological response (adjusting for age and sex). After two doses, 10 (53%) of 19 patients had CD4+ T-cell responses and 14 (74%) had CD8+ T-cell responses. A third vaccine dose induced serological responses in eight (16·3%) of 49 patients, but induced CD4+ and CD8+ T-cell responses in all patients assessed (n=12), including responses to the SARS-CoV-2 delta variant (B.1.617.2). Adverse events were reported in 32 (48%) of 67 patients and in 191 (78%) of 244 healthy controls after two doses, with the frequency not increasing after the third dose. There were no serious adverse events or deaths. INTERPRETATION This study provides important insight into the divergent humoral and cellular responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis. A third vaccine dose given 6-9 months after a rituximab infusion might not induce a serological response, but could be considered to boost the cellular immune response. FUNDING The Coalition for Epidemic Preparedness Innovations, Research Council of Norway Covid, the KG Jebsen Foundation, Oslo University Hospital, the University of Oslo, the South-Eastern Norway Regional Health Authority, Dr Trygve Gythfeldt og frues forskningsfond, the Karin Fossum Foundation, and the Research Foundation at Diakonhjemmet Hospital.
Collapse
Affiliation(s)
- Ingrid Jyssum
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hassen Kared
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Trung T Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Anne T Tveter
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Sella A Provan
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Joseph Sexton
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Kristin K Jørgensen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Grete B Kro
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - David J Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Eline B Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Tore K Kvien
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Ane Marie Anderson
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Espen A Haavardsholm
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | | | - Fridtjof Lund-Johansen
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig A Munthe
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Guro Løvik Goll
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
29
|
Habek M, Željko C, Savić Mlakar A, Bendelja K, Rogić D, Adamec I, Barun B, Gabelić T, Krbot Skorić M. Humoral and cellular immunity in convalescent and vaccinated COVID-19 people with multiple sclerosis: Effects of disease modifying therapies. Mult Scler Relat Disord 2022; 59:103682. [PMID: 35158189 PMCID: PMC8824161 DOI: 10.1016/j.msard.2022.103682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 01/09/2023]
Abstract
Objectives To determine anti-SARS-Cov2 antibodies and T-cell immunity in convalescent people with multiple sclerosis (pwMS) and/or pwMS vaccinated against Covid-19, depending on the disease modifying therapy, and in comparison to healthy controls (HC). Methods 75 participants were enrolled: Group 1—29 (38.7%) COVID-19 convalescent participants; Group 2—34 (45.3%) COVID-19 vaccinated; Group 3—12 (16.0%) COVID-19 convalescent participants who were later vaccinated against COVID-19. Cellular immunity was evaluated by determination of number of CD4+ and CD8+ cells secreting TNFα, IFNγ, and IL2 after stimulation with SARS-CoV-2 peptides. Results pwMS treated with ocrelizumab were less likely to develop humoral immunity after COVID-19 recovery or vaccination. No difference was observed in the cellular immunity in all studied parameters between pwMS treated with ocrelizumab compared to HC or pwMS who were treatment naïve or on first line therapies. These findings were consistent in convalescent, vaccinated, and convalescent+vaccinated participants. COVID-19 vaccinated convalescent pwMS on ocrelizumab compared to COVID-19 convalescent HC who were vaccinated did not show statistically difference in the rate of seroconversion nor titers of SARS-CoV-2 antibodies. Conclusion Presence of cellular immunity in pwMS on B-cell depleting therapies is reassuring, as at least partial protection from more severe COVID-19 outcomes can be expected.
Collapse
Affiliation(s)
- Mario Habek
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Cvetić Željko
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Ana Savić Mlakar
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Krešo Bendelja
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Dunja Rogić
- Clinical Institute for Laboratory Diagnostics, University Hospital Center Zagreb, Zagreb, Croatia
| | - Ivan Adamec
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Barbara Barun
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Tereza Gabelić
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Magdalena Krbot Skorić
- University Hospital Center Zagreb, Department of Neurology, Referral Center for Autonomic Nervous System Disorders, Zagreb, Croatia; Faculty of Electrical Engineering and Computing, University of Zagreb, Zagreb, Croatia
| |
Collapse
|