Multiple sclerosis disease-modifying therapies and COVID-19 vaccines: A practical review and meta-analysis ========================================================================================================== * Masoud Etemadifar * Hosein Nouri * Maristella Pitzalis * Maria Laura Idda * Mehri Salari * Mahshid Baratian * Sepide Mahdavi * Amir Parsa Abhari * Nahad Sedaghat ## Abstract **Importance** An evidence-based appraisal of the COVID-19 vaccination policies among people with multiple sclerosis (pwMS) with respect to disease-modifying therapies (DMT) is important for our understandings and their further management. **Objective** To synthesize the available evidence concerning the effect of DMTs on COVID-19 vaccination immunogenicity and effectiveness. **Data Sources** We searched MEDLINE, Scopus, Web of Science, MedRxiv, and Google Scholar from January 2021 until January 2022. **Study Selection** The exclusion criteria included: not a primary investigation; retracted/withdrawn; no eligible participants – people with no history/evidence of previous COVID-19 and corticosteroid administration within two months of vaccination; no eligible exposures – all nine DMT classes; and no eligible comparators – DMT-unexposed at the time of vaccination. **Data Extraction and Synthesis** Entries were assessed independently by two reviewers for eligibility and quality. Dichotomized data was extracted by two reviewers in accordance with Cochrane guidelines, and were pooled using either Peto fixed-effects or Inverse-variance random-effects methods. **Main Outcomes and Measures** Main outcomes were i) B-cell response, measured by seroconversion odds ratio (OR); ii) T-cell response, measured by interferon-gamma release response OR, and CD4+/CD8+ activation-induced marker+ OR. Further outcomes including immunity waning speed and breakthrough COVID-19 incidence/severity were synthesized narratively. **Results** Data from 28 studies (5,025 pwMS and 1,635 healthy participants) after COVID-19 vaccination suggests mildly-lower B-cell responses in teriflunomide- and alemtuzumab-treated, extensively-lower B-cell responses in sphingosine-1-phosphate receptor modulator (S1PRM)- and anti-CD20 (aCD20)-treated, and lower T-cell responses in interferon-, S1PRM-, alemtuzumab- and cladribine-treated pwMS. Every ten-week increase in aCD20-to-vaccine period is associated with a 1.94-time (95%CI: 1.57, 2.41, P<0.00001) increase in odds of seroconversion. B-cell-depleting therapies seem to accelerate post-vaccination humoral waning, and booster immunogenicity is predictable with the same factors affecting the priming vaccination. Furthermore, comparatively-increased breakthrough COVID-19 incidence and severity is being observed only among S1PRM- and anti-CD20-treated pwMS – i.e., among the pwMS with extensively-blunted B-cell response, despite adequate T-cell responses in the aCD20-treated. To date, pwMS on only-T-cell-blunting DMTs have not shown increased susceptibility to breakthrough COVID-19. **Conclusion and Relevance** The implemented vaccination strategy to date has been effective for pwMS on all DMTs other than S1PRM and aCD20. As B-cell immunity seems to be a more important predictor of vaccine effectiveness than T-cell immunity, optimization of humoral immunogenicity and ensuring its durability among pwMS on DMTs are the necessities of an effective COVID-19 vaccination policy. Key Words * multiple sclerosis * disease-modifying therapies * COVID-19 * vaccination * meta-analysis ## 1. Introduction From the beginning of the coronavirus disease 2019 (COVID-19), global mass vaccination has been the most prominent effort of humanity to end the reign of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Ever since, many vaccines have been developed, all with reasonable safety and efficacy profiles (more information available at: [https://covid19.trackvaccines.org/agency/who](https://covid19.trackvaccines.org/agency/who)). As the effectiveness of COVID-19 vaccination was thought to be altered in people with multiple sclerosis (pwMS) who receive immunomodulatory disease-modifying therapies (DMTs), expert panels issued adjusted vaccination guidelines for pwMS based on previous knowledge of the DMTs’ mechanisms of action and preliminary real-world evidence 1-4. These guidelines mostly concerned people on sphingosine 1-phosphate receptor modulators (S1PRM), anti-CD20 therapies (aCD20), and other B-cell depleting therapies (BCDT), as they were thought to blunt COVID-19 vaccines’ immunogenicity. Furthermore, administration of booster – in most cases, third – doses of COVID-19 vaccines was recommended after observation of waning humoral immunity 5 and clinical effectiveness 6-8. It was stressed after showing to be effective against the vaccine-escape 9 Omicron variant of the SARS-CoV-2 10-12 – regardless of the priming regimen 13,14. Among the pwMS, homologous booster doses were tested as a strategy to immunize the ones who did not seroconvert following their priming regimen. Now, several months after the mass vaccination of pwMS around the globe, the available real-world evidence seems adequate for a practical appraisal of the COVID-19 vaccination policies among pwMS with respect to their DMT. Hence, in this systematic review and meta-analysis study, we aimed to gather and synthesize the available evidence and highlight the gaps in the literature, providing a direction guide for future research, facilitating further policy makings, and enabling evidence-based management of pwMS. We hereby reported and discussed the results of our study (PROSPERO id: CRD42021278107) in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement (available from: [http://www.prisma-statement.org](http://www.prisma-statement.org)). The detailed methods of our study could be accessed from the online Supplementary Material. ## 2. Results and Discussion Overall, 28 studies including 5,025 pwMS and 1,635 healthy controls were synthesized (***Fig. 1, Table 1***). One study 15 was excluded despite containing eligible participants because their data could not be extracted. The studies differed in outcome measurement methods, settings, number of participants, and the administered vaccines. The assessed vaccines used either mRNA (BNT162b2, mRNA-1273), adenoviral vector (AV) (Ad26.COV2.S, ChAdOx1), or inactivated (CoronaVac, BBIBP-CorV) platforms. Other prominent points of heterogeneity were the used assays, the number/types of assessed DMTs, the usage of different comparators, e.g., healthy participants, pwMS on no DMT etc., and the timepoints of obtaining samples from participants. View this table: [Table 1.](http://medrxiv.org/content/early/2022/02/13/2022.02.12.22270883/T1) Table 1. Characteristics of included studies. ![Fig. 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/02/13/2022.02.12.22270883/F1.medium.gif) [Fig. 1.](http://medrxiv.org/content/early/2022/02/13/2022.02.12.22270883/F1) Fig. 1. PRISMA Flow Diagram. Abbreviation: pwMS, people with multiple sclerosis. The results of individual studies, heterogeneity tests, forest and funnel plots, and the detailed rationale behind each quality assessment – based on NIH tools – is accessible from the online Supplementary Material. The pooled measures are summarized in both ***Table 2 and Figure 2***, along with their certainty based on GRADE. View this table: [Table 2.](http://medrxiv.org/content/early/2022/02/13/2022.02.12.22270883/T2) Table 2. Summary of Findings. Results with statistically-significant P-values are bolded. ![Fig. 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/02/13/2022.02.12.22270883/F2.medium.gif) [Fig. 2.](http://medrxiv.org/content/early/2022/02/13/2022.02.12.22270883/F2) Fig. 2. Summary forest plot of the pooled results. Abbreviations: UX, unexposed; ref, reference; DMF, dimethyl fumarate; GA, glatiramer acetate; IFN, interferons; NTZ, natalizumab; CLAD, cladribine; TERI, teriflunomide; ALEM, alemtuzumab; aCD20, anti-CD20; S1PR, sphingosine-1-phosphate receptor modulators; IFN-g, interferon-gamma; AIM, activation-induced marker. ### 2.1. Effect of DMTs on COVID-19 vaccines’ immunogenicity #### 2.1.1. Interferons (IFN) Moderate-certainty evidence does not suggest decreased odds of post-vaccination seroconversion in the pwMS on IFNs compared to people unexposed (UX) to DMTs (OR [95%CI]: 0.84 [0.38, 1.83], P=0.66) (Supplementary Figure 1). No seronegative pwMS on IFN were present in four studies 16-19. Quantitative analysis in studies also did not suggest lower concentrations of antibodies among these pwMS post-vaccination. In one study, significantly higher concentrations of anti-Spike (S) receptor binding domain (RBD) IgG were detected among pwMS on IFNs, compared to healthy controls 19. Although the authors suggested that IFN-beta 1a therapy may promote the post-vaccination antibody responses in pwMS, this finding was not observed in other studies. Very-low-certainty evidence from one study 20 showed lower extents of interferon-gamma release response to the S antigen in samples from pwMS on IFNs, suggesting blunted T-cell response among these people compared to healthy controls (OR [95%CI]: 0.02 [0.00, 0.28], P<0.01). Both CD4+ and CD8+ T-cell responses were reduced in samples of pwMS on IFN compared to UX people, according to flow cytometric analysis in Tortorella et al. study 20. #### 2.1.2. Glatiramer Acetate (GA) Moderate-certainty evidence did not suggest decreased odds of post-vaccination seroconversion in the pwMS on GA compared to the UX people (OR [95%CI]: 0.87 [0.31, 2.42], P=0.79) (Supplementary Figure 2). Post-vaccination seronegative pwMS on GA were only present in two studies 21,22 – all being among the ones receiving inactivated vaccination. Quantitative analysis in suggested no difference in post-vaccination antibody concentrations between GA-treated pwMS and the UX people in any study. One study with limited sample size utilizing AIM assays 23 (very low-certainty evidence), suggested decreased odds of positive response neither in CD4+ (OR not measurable) nor CD8+ (OR [95%CI]: 0.62 [0.04, 9.00], P=0.72) T-cells. Furthermore, it is worth mentioning that adequate interferon-gamma release responses were present after SARS-CoV-2 *infection* in these people 24, suggesting favorable T-cell responses. #### 2.1.3. Dimethyl Fumarate (DMF) Moderate-certainty evidence did not suggest any decrease in odds of post-vaccination seroconversion among pwMS on DMF compared to UX people (OR [95%CI]: 1.98 [0.96, 4.09], P=0.07) (Supplementary Figure 3). Seven studies 16,18,19,22,23,25,26 did not contain any seronegative pwMS on DMF following mRNA or inactivated vaccination; Quantitative analysis suggested no difference in post-vaccination antibody concentrations compared to UX people. Similar to GA-treated pwMS, evidence on T-cell responses among these pwMS was limited to one study with a limited sample size 23 (very-low-certainty evidence), which by utilizing AIM assays, suggested no decrease in odds of positive responses in subsets of CD4+ (OR not measurable) and CD8+ (OR [95%CI]: 3.78 [0.18, 78.38], P=0.39) T-cells. Similarly, interferon-gamma release responses were sufficient in DMF-treated pwMS after SARS-CoV-2 *infection*, suggesting adequate T-cell response 24. #### 2.1.4. Teriflunomide (TERI) Inadequate number of studies with considerable heterogeneity (very-low-certainty evidence) suggest decreased odds of post-vaccination seroconversion in pwMS on TERI compared to UX people (OR [95%CI]: 0.38 [0.16, 0.90], P=0.03) (Supplementary Figure 4). Due to low number of mRNA vaccine studies with measurable relative effect and therefore, the uncertainty of their pooled measures, the difference between the inactivated and mRNA measures did not reach statistical significance (Chi2=2.91, P=0.09). Nevertheless, considering that none of the TERI-treated pwMS in any of the studies 16,18,22,25-28 remained seronegative after mRNA vaccination, the suggested blunt caused by TERI may not be generalizable to the mRNA-vaccinated pwMS. Furthermore, lower antibody concentrations compared to UX people were observed in pwMS on TERI homogenously in all studies regardless of the used vaccine, but reached statistical significance only in one 28. TERI’s mechanism of action – which involves inhibition of rapidly-dividing cells, including activated B-cells – may explain this observation. Furthermore, no evidence was found regarding the T-cell responses in these pwMS following vaccination; Assessing the vaccine-induced T-cell responses in TERI-treated pwMS is therefore encouraged. #### 2.1.5. Sphingosine-1-phosphate receptor modulators (S1PRM) High-certainty evidence confirms significantly lower odds of post-vaccination seroconversion in pwMS on S1PRM compared with UX people (OR [95%CI]: 0.04 [0.03, 0.06], P<0.00001) (Supplementary Figure 5). All included studies 16,17,19-22,25-31 found significantly lower concentrations of antibodies following vaccination in these pwMS compared to UX people. Studies with heterogenous effect measures (moderate-certainty evidence) indicate that with the current vaccination strategy, pwMS on S1PRM are 25 times (95%CI: 16.66, 33.33) less likely to show anti-S1, and 8.33 times (95%CI: 3.70, 20) less likely to show anti-S seroconversion following COVID-19 vaccination (Chi2=7.24, P<0.01). Paradoxical to the healthy population, low-certainty evidence – due to limited count of inactivated vaccine studies – suggests that among the pwMS on S1PRM, odds of anti-S1 seroconversion is higher with inactivated vaccines compared to mRNA and AV vaccines (Chi2=11.97, P<0.001). Although theoretically reasonable 32, head-to-head mRNA-AV 29 and mRNA-inactivated 22 immunogenicity comparisons in S1PRM-treated pwMS have suggested the opposite. Hence, the need for more replication of inactivated/mRNA/AV comparisons is clearly sensed for pwMS on S1PRM. Furthermore, interferon-gamma release assays in two studies 20,26 (low-certainty evidence) suggested decreased odds of positive T-cell response in pwMS on S1PRM (OR [95%CI]: 0.04 [0.02, 0.07], P<0.00001) (Supplementary Figure 6). AIM assay in another study 23 (very-low-certainty evidence) did not suggest decreased odds of CD8+ T-cell response in these pwMS (OR [95%CI]: 0.95 [0.08, 10.71], P=0.97) but suggested decreased odds of CD4+ T-cell responses (OR [95%CI]: 0.01 [0.00, 0.18], P=0.001) compared to UX people. Additionally, among the pwMS on S1PRM who failed to seroconvert following priming vaccination, one study showed that administration of booster doses increased anti-S1 antibody concentrations, but promoted seroconversion only in 2/29 (7%) 33. The trafficking inhibition of lymphocytes, their restriction to lymphatics, and hence, the peripheral lymphopenia seen in pwMS on S1PRM explains the lower T-cell reactivity observed in peripheral blood samples – not to mention S1PRM’s documented inhibitive effect on T-cell activation 34. The former reason may also explain the blunted humoral responses, as the one-way flow of lymph from peripheral to central areas restricts trafficking of lymphocytes to peripheral areas, inhibiting proper exposure of lymph-trapped lymphocytes to immunizing materials of the vaccines which are administered peripherally/locally. As systemic SARS-CoV-2 infection mounts adequate immunization among pwMS on S1PRM 32,35, it can be hypothesized that immunogenicity in these people is subject to wider (e.g., systemic instead of local/peripheral) exposure to immunogens. When the lymphocytes’ ability to reach the immunogens is inhibited, the immunogens should reach the lymphocytes themselves or immunization will not develop. #### 2.1.6. Natalizumab (NTZ) Compared to UX people, low-certainty evidence did not confirm lower odds of anti-S1 seroconversion among pwMS on NTZ following vaccination (OR [95%CI]: 0.53 [0.24, 1.18]) (Supplementary Figure 7). All pwMS on NTZ in seven studies – including the only two studies using anti-S assays with extractable data 23,36 – seroconverted following vaccination 16,17,23,25,26,28,36. Although relative effect was measurable in only two of them which contained seronegative UX people 26,28. Unlike all the other studies, pwMS on NTZ in one study 28 showed significantly lower post-vaccination antibodies compared to UX people. Interferon-gamma release 17 and AIM 23 assays (very low-certainty evidence) did not suggest blunted post-vaccination T-cell responses in pwMS on NTZ compared to UX people. NTZ, an anti-α4-integrin monoclonal antibody, implements its effect by inhibiting lymphocyte extravasation; however, unlike S1PRM, its lymphocyte trafficking inhibition does not trap the lymphocytes in the lymphatic system – i.e., it does not cause peripheral lymphopenia. Although their trafficking abilities are inhibited, the preserved presence of lymphocytes in blood flow – which, unlike the lymph flow, can be from central to peripheral areas as well – may be the reason NTZ does not blunt vaccination-induced immunization as much as S1PRM. #### 2.1.7. Cladribine (CLAD) Pooled low-certainty evidence (Supplementary Figure 8) confirms no difference in odds of anti-S1 seroconversion among pwMS on CLAD compared to UX people (OR [95%CI]: 0.41 [0.15, 1.11], P=0.08). No evidence was found regarding anti-S seroconversion. In five studies 16,19,20,22,25, all CLAD-treated pwMS seroconverted following vaccination similar to the UX people, preventing relative effect measurement. Quantitative analysis in no studies suggested lower concentrations of post-vaccination antibodies among them. Assessment of vaccination-induced T-cell response was limited to one study 20 (very-low-certainty evidence); it showed lower odds of positive S-induced interferon-gamma release responses in samples from pwMS on CLAD compared to UX people (OR [95%CI]: 0.01 [0.00, 0.04], P<0.00001). Compared to its effect on the T-cell lineage, CLAD’s effect on the B-cells is more extensive but less durable 37-40. As interpreted, this has been translated into observation of proper humoral despite blunted cellular immunization following COVID-19 vaccination among pwMS on CLAD. Furthermore, the time since the last CLAD dose theoretically affects humoral responses; This was suggested especially by Achiron et al. study 26 but did not reach statistical significance, and was not confirmed by other studies 25,28,41; It seems the implemented guidelines 1-4 have suggested an adequate amount of post-CLAD vaccination delay to prevent blunted humoral responses, and therefore, made the probable effect unmeasurable. Additionally, although CLAD depletes the memory B-cells 42, a preliminary study suggested its subsequent doses will not alter pre-existing humoral memory 43; Still, there is limited evidence that the longevity of COVID-19 vaccine-induced humoral responses is lower in pwMS on CLAD 26. Replicative studies measuring the immunity waning speed in these pwMS after COVID-19 vaccination are, therefore, required to determine whether they require personalized booster schedules. #### 2.1.8. Alemtuzumab (ALEM) Pooled very-low-certainty evidence (Supplementary Figure 9) suggests lower odds of anti-S1 seroconversion among pwMS on ALEM compared to UX people (OR [95%CI]: 0.32 [0.10, 0.96], P=0.04). One study assessed anti-S seroconversion, but its data could not be extracted 30. PwMS on ALEM in three studies 16,25,28 showed 100% seroconversion rates similar to the UX people in two of them 16,25, and none of the studies suggested lower concentrations of post-vaccination antibodies among them – not even the study that indicated lower odds of seroconversion 26. Vaccination-induced T-cell responses were not assessed in any of the included studies, still, ALEM’s durable effect of T-cell lineage 44 suggests that they are blunted . ALEM, an anti-CD52 monoclonal antibody, is known to significantly deplete B- and T-cells shortly after administration. ALEM’s short-term effect on B- and T-cell dynamics is relatively similar to CLAD 40,44. Hence, although the time from the last ALEM infusion affects seroconversion 26, this effect is currently not measurable as the implemented guidelines 1-4 seem to have suggested an adequate amount of delay. Similar to other BCDT, further studies measuring comparative immunity waning speeds in pwMS on ALEM are needed to determine whether they require more frequent boosters. #### 2.1.9. Anti-CD20 therapies (aCD20) High-certainty evidence confirms lower odds of seroconversion following COVID-19 vaccination among pwMS on aCD20 compared to UX people (OR [95%CI]: 0.05 [0.04, 0.06], P<0.00001) (Supplementary Figure 10). Studies with heterogenous effect measures (moderate-certainty evidence) indicate that with the currently-implemented strategies, pwMS on aCD20 are 20 times (95%CI: 16.66, 25) less likely to seroconvert for anti-S1, and 12.5 times (95%CI: 7.69, 20) less likely to seroconvert for anti-S antibodies (Chi2=2.76, P=0.10) following COVID-19 vaccination. Quantitative analyses in all included studies confirmed this observation. Furthermore, evidence indicates with high certainty that every 10-week delay in subsequent aCD20 infusion is associated with a 1.94-time (95%CI: 1.57, 2.41, P<0.00001) increase in seroconversion odds of pwMS on aCD20 (Supplementary Figure n). Regarding the T-cell responses, compared to UX people, moderate-certainty evidence does not suggest different odds of positive post-vaccination T-cell interferon-gamma release responses (OR [95%CI]: 1.12 [0.62, 2.05], P=0.70) (Supplementary Figure 12), CD8+ (OR [95%CI]: 2.54 [0.89, 7.27], P=0.08) (Supplementary Figure 13), and CD4+ (OR [95%CI]: 1.13 [0.17, 7.61], P=0.90) T-cell AIM responses. Quantitative analyses in most studies 17,20,23,29,45-48 were in line with the dichotomized evidence. Multiplex polymerase chain reaction assay in one study 49 indicated positive adaptive T-cell responses among 100% of seronegative pwMS on aCD20 following vaccination. Furthermore, the preliminary evidence indicates significant decline in seropositivity rates of pwMS on aCD20 six months after their second dose 50,51. Homologous mRNA boosters in pwMS on aCD20 promoted T-cell responses 52, while humoral responses were still heavily dependent on the serostatus following the priming regimen, and B-cell dynamics at the time of booster administration 33,51-53; In other words, the booster doses did not promote humoral immunization in pwMS on aCD20 who did not seroconvert following priming vaccination, unless their B-cells were reconstituted. Studies among people on aCD20 with diseases other than MS 54,55 support the same conclusion. Similar to pwMS on other DMTs, the COVID-19 vaccines’ immunogenicity among pwMS on aCD20 could be considered a translation of the previously-determined B- and T-cell dynamics in them 40,56, based on which the current guidelines recommended a 12-to-36-week window between aCD20 infusion and COVID-19 vaccination 1-4. However, the presented evidence suggests that the mentioned interval, although increases the odds, will not be adequate to reverse the humoral blunts in pwMS on aCD20 (Fig. 3). The alterations in the dynamics of B-cells in people receiving aCD20 last for years according to the unpublished results from the [NCT00676715](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT00676715&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) phase-II extension trial 57, suggesting durable, long-lasting benefits of aCD20 without subsequent dosing 58. However, this durable effect of aCD20 has shown to be able to alter vaccine immunogenicity for as long as three years, as observed in people with hematological malignancies 59. Hence, prior B-cell profiling and post-vaccination serological screening may be the necessities of an effective personalized vaccination strategy in pwMS who received aCD20 at any time point within three years. ![Fig. 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/02/13/2022.02.12.22270883/F3.medium.gif) [Fig. 3.](http://medrxiv.org/content/early/2022/02/13/2022.02.12.22270883/F3) Fig. 3. Schematic curve showing the association of post-vaccination seroconversion rates with time since last anti-CD20 infusion. *Current guideline recommendations on minimum delay of vaccination after anti-CD20 infusion. Abbreviations: UX, unexposed; aCD20, anti-CD20. ### 2.2. Vaccine types Based on phase-III data, the efficacy profiles of different available COVID-19 vaccine types (i.e., mRNA, AV, inactivated, and protein-based) seems to correlate to their anti-S/S1 humoral immunogenicity – both in healthy people 60,61 and pwMS 62. Head-to-head comparisons of the available COVID-19 vaccines’ humoral immunogenicity among pwMS reveals the superiority of mRNA-1237 over BNT162b2 (both mRNA-based) 20,25,63 – probably due to higher concentrations of active material, BNT162b2 and mRNA-1237 (mRNA) over ChAdOx1 and Ad26.COV2 (AV) 17,23,29,49 and BNT162b2 (mRNA) over CoronaVac (inactivated) 22 – although humoral immunization did not differ significantly in pwMS on aCD20 receiving BNT162b2 and CoronaVac in Ozakbas et al. 22 study. The choice of a specific vaccine type among pwMS is encouraged and should be based on an individualized risk/benefit assessment with careful consideration of their COVID-19 risk factor profile 64,65, their DMT, and the availability/cost-effectiveness of the vaccine 66,67. ### 2.3. Moving into the clinic While the serum anti-S/S1 assays are deemed predictive of its neutralizing activity 68,69, and the serum neutralizing activity predictive of the clinical protection against symptomatic SARS-CoV-2 infection 70, pwMS with adequate humoral responses – mostly on DMTs other than aCD20 and S1PRM – would theoretically show adequate protection against SARS-CoV-2. However, in the absence of humoral immunization, it is doubted if the T-cells could provide adequate clinical protection among pwMS on aCD20 and S1PRM. The current real-world evidence confirms the predictive effect of seroconversion on post-vaccination COVID-19 incidence and severity 62, and indicates rising comparative incidence and severity of COVID-19 among pwMS on S1PRM and aCD20 following vaccination of pwMS 62,71-73. The less-extensive humoral blunts in pwMS on TERI – and possibly ALEM – do not seem to have had any significant effect on vaccine effectiveness. Regarding the protective effect of T-cell responses, currently, the only clue lies within the Etemadifar et al. 72 study – although not confirmed by a larger study 73; it showed that among the vaccinated pwMS, the ones on aCD20 experienced lower incidence and severity of COVID-19 in comparison with those on fingolimod. The former are known to be prone to worse COVID-19 outcomes 64 but show robust post-vaccination T-cell responses, while the latter do not develop proper T-cell immunization following vaccination. Still, the practical protective effect of T-cell responses in the absence of antibodies could neither be confirmed nor measured until further real-world evidence becomes available. ## 3. Conclusion The present analysis highlight and corroborate the relevance for an optimal treatment strategy in pwMS before COVID-19 vaccination. It was demonstrated that the current vaccination strategy has failed to promote adequate humoral immunity in aCD20-and S1PRM-treated pwMS, which is being translated into low clinical effectiveness of COVID-19 vaccines among them – despite adequate T-cell responses in the ones on aCD20. Their susceptibility to worse COVID-19 outcomes, and the dependency of COVID-19 vaccines’ humoral immunogenicity to the B-cell dynamics at the time of administration – and therefore, the timing of aCD20 infusion – stress the importance of personalizing vaccination strategies for pwMS on aCD20 with respect to their B-cell profiles and aCD20 infusion timings. Theoretically and based on limited evidence, mode of action and administration method may be important factors to consider also when vaccinating S1PR modulator-treated pwMS, while more evidence is needed to support this claim. Milder humoral and considerable T-cell response blunts – also depending on dosage timings, and higher immunity waning speeds may be present in pwMS on CLAD and ALEM, which subject to confirmation by further evidence, stresses the importance of earlier booster administrations among them. TERI may also cause a humoral immunogenicity blunt, however, being less extensive and clinically-irrelevant based on current evidence; PwMS on TERI may not require countering policies other than being provided with reliable information about the importance of booster doses. Evidence to date does not indicate any significant effect of IFN, GA, DMF, and NTZ on COVID-19 vaccines’ immunogenicity and effectiveness. Additionally, as heterologous boosters among healthy people showed to be more immunogenic and effective 74, further replication among pwMS with heterologous regimens is encouraged, especially among the ones primed with inactivated vaccines – as samples from inactivated-vaccinated people show less neutralizing activity against the Omicron variant 75, and the heterologous boosters with superiority of mRNA 13,14,76-78 over AV 79 have shown to be more immunogenic than homologous boosters in healthy people receiving inactivated priming regimens. ## Supporting information Methods and Supplementary Figures [[supplements/270883_file05.docx]](pending:yes) Quality Assessment [[supplements/270883_file06.docx]](pending:yes) ## Data Availability Not applicable. This work is a secondary study and no data was produced. ## 5. Conflict of Interest and Funding The authors declare no conflict of interest. This study did not receive any funding. ## 4. Acknowledgements We would like to thank Prof. Mohammad Reza Maracy for his valuable methodological consultations. * Received February 12, 2022. * Revision received February 12, 2022. * Accepted February 13, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## 6. References 1. 1.Timing MS Medications with COVID-19 Vaccines. National MS Society. Accessed 19 November 2021. [https://www.nationalmssociety.org/coronavirus-covid-19-information/multiple-sclerosis-and-coronavirus/covid-19-vaccine-guidance/Timing-MS-Medications-with-COVID-19-Vaccines](https://www.nationalmssociety.org/coronavirus-covid-19-information/multiple-sclerosis-and-coronavirus/covid-19-vaccine-guidance/Timing-MS-Medications-with-COVID-19-Vaccines) 2. 2.Yamout BI, Zakaria M, Inshasi J, et al. MENACTRIMS practice guideline for COVID-19 vaccination in patients with multiple sclerosis. Multiple Sclerosis and Related Disorders. 2021;56:103225. 3. 3.MS, DMTs and COVID-19 vaccines consensus statement. Multiple Sclerosis Society. Accessed 13 January, 2022. [https://www.mssociety.org.uk/what-we-do/news/ms-society-medical-advisers-release-consensus-statement-covid-19-vaccines](https://www.mssociety.org.uk/what-we-do/news/ms-society-medical-advisers-release-consensus-statement-covid-19-vaccines) 4. 4.Wolf A, Alvarez E. COVID-19 Vaccination in Patients With Multiple Sclerosis on Disease-Modifying Therapy. Neurology: Clinical Practice. 2021; 5. 5.Levin EG, Lustig Y, Cohen C, et al. Waning immune humoral response to BNT162b2 Covid-19 vaccine over 6 months. New England Journal of Medicine. 2021;385(24):e84. 6. 6.Goldberg Y, Mandel M, Bar-On YM, et al. Waning immunity after the BNT162b2 vaccine in Israel. New England Journal of Medicine. 2021;385(24):e85. 7. 7.Rosenberg ES, Dorabawila V, Easton D, et al. Covid-19 Vaccine Effectiveness in New York State. N Engl J Med. Jan 13 2022;386(2):116–127. doi:10.1056/NEJMoa2116063 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2116063&link_type=DOI) 8. 8.Lin DY, Gu Y, Wheeler B, et al. Effectiveness of Covid-19 Vaccines over a 9-Month Period in North Carolina. N Engl J Med. Jan 12 2022;doi:10.1056/NEJMoa2117128 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2117128&link_type=DOI) 9. 9.Edara VV, Manning KE, Ellis M, et al. mRNA-1273 and BNT162b2 mRNA vaccines have reduced neutralizing activity against the SARS-CoV-2 Omicron variant. bioRxiv. Dec 22 2021;doi:10.1101/2021.12.20.473557 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4xMi4yMC40NzM1NTd2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAyLzEzLzIwMjIuMDIuMTIuMjIyNzA4ODMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 10. 10.Accorsi EK, Britton A, Fleming-Dutra KE, et al. Association Between 3 Doses of mRNA COVID-19 Vaccine and Symptomatic Infection Caused by the SARS-CoV-2 Omicron and Delta Variants. Jama. Jan 21 2022;doi:10.1001/jama.2022.0470 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2022.0470&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35060999&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 11. 11.Gruell H, Vanshylla K, Tober-Lau P, et al. mRNA booster immunization elicits potent neutralizing serum activity against the SARS-CoV-2 Omicron variant. Nat Med. Jan 19 2022:1-4. doi:10.1038/s41591-021-01676-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01676-0&link_type=DOI) 12. 12.Garcia-Beltran WF, St Denis KJ, Hoelzemer A, et al. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell. Jan 6 2022;doi:10.1016/j.cell.2021.12.033 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.12.033&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34995482&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 13. 13.Pérez-Then E, Lucas C, Monteiro VS, et al. Neutralizing antibodies against the SARS-CoV-2 Delta and Omicron variants following heterologous CoronaVac plus BNT162b2 booster vaccination. Nat Med. Jan 20 2022;doi:10.1038/s41591-022-01705-6 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-01705-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35051990&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 14. 14.Cheng SMS, Mok CKP, Leung YWY, et al. Neutralizing antibodies against the SARS-CoV-2 Omicron variant following homologous and heterologous CoronaVac or BNT162b2 vaccination. Nat Med. Jan 20 2022;doi:10.1038/s41591-022-01704-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-01704-7&link_type=DOI) 15. 15.Bigaut K, Kremer L, Fleury M, Lanotte L, Collongues N, de Seze J. Impact of disease-modifying treatments on humoral response after COVID-19 vaccination: a mirror of the response after SARS-CoV-2 infection. Revue Neurologique. 2021; 16. 16.Capone F, Lucchini M, Ferraro E, et al. Immunogenicity and safety of mRNA COVID-19 vaccines in people with multiple sclerosis treated with different disease-modifying therapies. Neurotherapeutics. 2021:1–9. 17. 17.Gadani SP, Reyes-Mantilla M, Jank L, et al. Discordant humoral and T cell immune responses to SARS-CoV-2 vaccination in people with multiple sclerosis on anti-CD20 therapy. EBioMedicine. 2021;73:103636. 18. 18.Giossi R, Consonni A, Clerici VT, et al. Anti-Spike IgG in multiple sclerosis patients after BNT162b2 vaccine: An exploratory case-control study in Italy. Multiple Sclerosis and Related Disorders. 2021:103415. 19. 19.Maniscalco GT, Manzo V, Ferrara AL, et al. Interferon Beta-1a treatment promotes SARS-CoV-2 mRNA vaccine response in multiple sclerosis subjects. Multiple sclerosis and related disorders. 2022;58:103455. 20. 20.Tortorella C, Aiello A, Gasperini C, et al. Humoral-and T-Cell – Specific Immune Responses to SARS-CoV-2 mRNA Vaccination in Patients With MS Using Different Disease-Modifying Therapies. Neurology. 2021; 21. 21.Etemadifar M, Sedaghat N, Nouri H, et al. SARS-CoV-2 serology among people with multiple sclerosis on disease-modifying therapies after BBIBP-CorV (Sinopharm) inactivated virus vaccination: Same story, different vaccine. Multiple Sclerosis and Related Disorders. 2022;57:103417. 22. 22.Ozakbas S, Baba C, Dogan Y, Cevik S, Ozcelik S, Kaya E. Comparison of SARS-CoV-2 antibody response after two doses of mRNA and inactivated vaccines in multiple sclerosis patients treated with disease-modifying therapies. Multiple sclerosis and related disorders. 2022:103486. 23. 23.Sabatino Jr JJ, Mittl K, Rowles W, et al. Impact of multiple sclerosis disease-modifying therapies on SARS-CoV-2 vaccine-induced antibody and T cell immunity. medRxiv. 2021; 24. 24.Kister I, Patskovsky Y, Curtin R, et al. 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. medRxiv. 2022; 25. 25.Sormani MP, Inglese M, Schiavetti I, et al. Effect of SARS-CoV-2 mRNA vaccination in MS patients treated with disease modifying therapies. EBioMedicine. 2021;72:103581. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 26. 26.Achiron A, Mandel M, Dreyer-Alster S, et al. Humoral immune response in multiple sclerosis patients following PfizerBNT162b2 COVID19 vaccination: Up to 6 months cross-sectional study. Journal of Neuroimmunology. 2021;361:577746. 27. 27.Disanto G, Sacco R, Bernasconi E, et al. Association of disease-modifying treatment and anti-CD20 infusion timing with humoral response to 2 SARS-CoV-2 vaccines in patients with multiple sclerosis. JAMA neurology. 2021;78(12):1529–1531. 28. 28.Pitzalis M, Idda ML, Lodde V, et al. Effect of different disease-modifying therapies on humoral response to BNT162b2 vaccine in Sardinian multiple sclerosis patients. Frontiers in immunology. 2021:5233. 29. 29.Tallantyre EC, Vickaryous N, Anderson V, et al. COVID-19 Vaccine Response in People with Multiple Sclerosis. Annals of neurology. 2022;91(1):89–100. 30. 30.König M, Lorentzen ÅR, Torgauten HM, et al. Humoral immunity to SARS-CoV-2 mRNA vaccination in multiple sclerosis: the relevance of time since last rituximab infusion and first experience from sporadic revaccinations. Journal of Neurology, Neurosurgery & Psychiatry. 2021; 31. 31.Türkoğlu R, Baliç N, Kızılay T, et al. 7Fingolimod impairs inactivated vaccine (CoronaVac)-induced antibody response to SARS-CoV-2 spike protein in persons with multiple sclerosis. Multiple Sclerosis and Related Disorders. 2022:103524. 32. 32.Rommer PS, Bsteh G, Berger T, Zettl UK. SARS-CoV-2 antibodies in multiple sclerosis patients depending on the vaccine mode of action? Multiple Sclerosis Journal. 2022;28(1):165–167. 33. 33.König M, Torgauten HM, Øverås MH, et al. Efficacy and safety of a third SARS-CoV-2 vaccination in multiple sclerosis vaccine non-responders. medRxiv. 2021:2021.10.15.21264977. doi:10.1101/2021.10.15.21264977 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4xMC4xNS4yMTI2NDk3N3YxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDIvMTMvMjAyMi4wMi4xMi4yMjI3MDg4My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. 34.Baer A, Colon-Moran W, Bhattarai N. Characterization of the effects of immunomodulatory drug fingolimod (FTY720) on human T cell receptor signaling pathways. Scientific Reports. 2018/07/19 2018;8(1):10910. doi:10.1038/s41598-018-29355-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-018-29355-0&link_type=DOI) 35. 35.Sormani MP, Schiavetti I, Landi D, et al. SARS-CoV-2 serology after COVID-19 in multiple sclerosis: An international cohort study. Multiple Sclerosis Journal. 2021:13524585211035318. 36. 36.Capuano R, Donnarumma G, Bisecco A, et al. Humoral response to SARS-CoV-2 mRNA vaccine in patients with multiple sclerosis treated with natalizumab. Therapeutic Advances in Neurological Disorders. 2021;14:17562864211038111. 37. 37.Giovannoni G, Comi G, Cook S, et al. A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis. New England Journal of Medicine. 2010;362(5):416–426. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa0902533&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20089960&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000274193300007&link_type=ISI) 38. 38.Duddy M, Niino M, Adatia F, et al. Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis. The Journal of Immunology. 2007;178(10):6092–6099. 39. 39.Comi G, Cook S, Giovannoni G, et al. Effect of cladribine tablets on lymphocyte reduction and repopulation dynamics in patients with relapsing multiple sclerosis. Multiple sclerosis and related disorders. 2019;29:168–174. 40. 40.Baker D, MacDougall A, Kang AS, Schmierer K, Giovannoni G, Dobson R. CD19 B cell repopulation after ocrelizumab, alemtuzumab and cladribine: Implications for SARS-CoV-2 vaccinations in multiple sclerosis. Multiple Sclerosis and Related Disorders. 2022;57:103448. 41. 41.Grothe C, Steffen F, Bittner S. Humoral immune response and lymphocyte levels after complete vaccination against COVID-19 in a cohort of multiple sclerosis patients treated with cladribine tablets. Journal of Central Nervous System Disease. 2021;13:11795735211060118. 42. 42.Baker D, Marta M, Pryce G, Giovannoni G, Schmierer K. Memory B cells are major targets for effective immunotherapy in relapsing multiple sclerosis. EBioMedicine. 2017;16:41–50. 43. 43.Moser T, O’Sullivan C, Puttinger C, et al. Pre-Existing Humoral Immunological Memory Is Retained in Patients with Multiple Sclerosis Receiving Cladribine Therapy. Biomedicines. 2021;9(11):1584. 44. 44.Ruck T, Bittner S, Wiendl H, Meuth SG. Alemtuzumab in multiple sclerosis: mechanism of action and beyond. International journal of molecular sciences. 2015;16(7):16414–16439. 45. 45.Apostolidis SA, Kakara M, Painter MM, et al. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nature medicine. 2021;27(11):1990–2001. 46. 46.Madelon N, Lauper K, Breville G, et al. Robust T cell responses in anti-CD20 treated patients following COVID-19 vaccination: a prospective cohort study. Clinical Infectious Diseases. 2021; 47. 47.Brill L, Rechtman A, Zveik O, et al. Humoral and T-cell response to SARS-CoV-2 vaccination in patients with multiple sclerosis treated with ocrelizumab. JAMA neurology. 2021;78(12):1510–1514. 48. 48.Pompsch M, Fisenkci N, Horn PA, Kraemer M, Lindemann M. Evidence of extensive cellular immune response after SARS-CoV-2 vaccination in ocrelizumab-treated patients with multiple sclerosis. Neurological Research and Practice. 2021;3(1):1–6. 49. 49.Katz J, Bouley A, Jungquist R, Douglas E, O’Shea I, Lathi E. Humoral and T-cell responses to SARS-CoV-2 vaccination in multiple sclerosis patients treated with ocrelizumab. Multiple Sclerosis and Related Disorders. 2021:103382. 50. 50.Bajwa HM, Novak F, Nilsson AC, et al. Persistently reduced humoral and cellular immune response following third SARS-CoV-2 mRNA vaccination in anti-CD20-treated multiple sclerosis patients. medRxiv. 2022; 51. 51.Brill L, Raposo C, Rechtman A, et al. SARS-CoV-2 third vaccine immune response in MS patients treated with ocrelizumab. medRxiv. 2022; 52. 52.Madelon N, Heikkilä N, Sabater Royo I, et al. Omicron-specific cytotoxic T-cell responses are boosted following a third dose of mRNA COVID-19 vaccine in anti-CD20-treated multiple sclerosis patients. medRxiv. 2021:2021.12.20.21268128. doi:10.1101/2021.12.20.21268128 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4xMi4yMC4yMTI2ODEyOHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDIvMTMvMjAyMi4wMi4xMi4yMjI3MDg4My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 53. 53.Achtnichts L, Jakopp B, Oberle M, et al. Humoral Immune Response after the Third SARS-CoV-2 mRNA Vaccination in CD20 Depleted People with Multiple Sclerosis. Vaccines. 2021;9(12):1470. 54. 54.Felten R, Gallais F, Schleiss C, et al. Cellular and humoral immunity after the third dose of SARS-CoV-2 vaccine in patients treated with rituximab. Lancet Rheumatol. Jan 2022;4(1):e13–e16. doi:10.1016/s2665-9913(21)00351-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s2665-9913(21)00351-9&link_type=DOI) 55. 55.Bonelli M, Mrak D, Tobudic S, et al. Additional heterologous versus homologous booster vaccination in immunosuppressed patients without SARS-CoV-2 antibody seroconversion after primary mRNA vaccination: a randomised controlled trial. Ann Rheum Dis. Jan 13 2022;doi:10.1136/annrheumdis-2021-221558 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjI1OiJhbm5yaGV1bWRpcy0yMDIxLTIyMTU1OHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDIvMTMvMjAyMi4wMi4xMi4yMjI3MDg4My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 56. 56.Kappos L, Li D, Calabresi PA, et al. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. The Lancet. 2011;378(9805):1779–1787. 57. 57.Hauser S, Li D, Calabresi P, et al. Week 144 results of a phase II, randomized, multicenter trial assessing the safety and efficacy of ocrelizumab in patients with relapsing – remitting multiple sclerosis (RRMS)(S31. 004). AAN Enterprises; 2013. 58. 58.Baker D, Pryce G, James LK, Marta M, Schmierer K. The ocrelizumab phase II extension trial suggests the potential to improve the risk: Benefit balance in multiple sclerosis. Multiple Sclerosis and Related Disorders. 2020;44:102279. 59. 59.Funakoshi Y, Yakushijin K, Ohji G, et al. Increase in Antibody Titers Following Sars-Cov-2 Vaccination Remains Limited for More Than 3 Years after Final Dose of Anti-CD20 Antibody. Blood. 2021;138:534. 60. 60.Fan Y-J, Chan K-H, Hung IF-N. Safety and Efficacy of COVID-19 Vaccines: A Systematic Review and Meta-Analysis of Different Vaccines at Phase 3. Vaccines. 2021;9(9):989. 61. 61.Cheng H, Peng Z, Luo W, et al. Efficacy and Safety of COVID-19 Vaccines in Phase III Trials: A Meta-Analysis. Vaccines. 2021;9(6):582. 62. 62.Sormani MP, Schiavetti I, Inglese M, et al. Breakthrough SARS-CoV-2 infections after COVID-19 mRNA vaccination in MS patients on disease modifying therapies. medRxiv. 2021; 63. 63.Ali A, Dwyer D, Wu Q, et al. Characterization of humoral response to COVID mRNA vaccines in multiple sclerosis patients on disease modifying therapies. Vaccine. 2021;39(41):6111–6116. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2021.08.078&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34483021&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 64. 64.Etemadifar M, Nouri H, Maracy MR, et al. Risk factors of severe COVID-19 in people with multiple sclerosis: A systematic review and meta-analysis. Revue neurologique. 2021; 65. 65.Sormani MP, Schiavetti I, Carmisciano L, et al. COVID-19 severity in multiple sclerosis: putting data into context. Neurology-Neuroimmunology Neuroinflammation. 2022;9(1) 66. 66.López F, Català M, Prats C, et al. A Cost-Benefit Analysis of COVID-19 Vaccination in Catalonia. Vaccines (Basel). Dec 31 2021;10(1)doi:10.3390/vaccines10010059 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/vaccines10010059&link_type=DOI) 67. 67.Vaezi A, Meysamie A. COVID-19 Vaccines Cost-Effectiveness Analysis: A Scenario for Iran. Vaccines (Basel). Dec 29 2021;10(1)doi:10.3390/vaccines10010037 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/vaccines10010037&link_type=DOI) 68. 68.Kohmer N, Westhaus S, Rühl C, Ciesek S, Rabenau HF. Brief clinical evaluation of six high-throughput SARS-CoV-2 IgG antibody assays. Journal of Clinical Virology. 2020/08/01/ 2020;129:104480. doi:[https://doi.org/10.1016/j.jcv.2020.104480](https://doi.org/10.1016/j.jcv.2020.104480) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jcv.2020.104480&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32505777&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 69. 69.Müller L, Ostermann PN, Walker A, et al. Sensitivity of anti-SARS-CoV-2 serological assays in a high-prevalence setting. European Journal of Clinical Microbiology & Infectious Diseases. 2021/05/01 2021;40(5):1063–1071. doi:10.1007/s10096-021-04169-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10096-021-04169-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33534090&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 70. 70.Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nature Medicine. 2021/07/01 2021;27(7):1205–1211. doi:10.1038/s41591-021-01377-8 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01377-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34002089&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 71. 71.Garjani A, Patel S, Bharkhada D, et al. Impact of mass vaccination on SARS-CoV-2 infections among multiple sclerosis patients taking immunomodulatory disease-modifying therapies in England. Multiple Sclerosis and Related Disorders. 2022;57:103458. 72. 72.Etemadifar M, Abhari AP, Nouri H, Eighani N, Salari M, Sedaghat N. Effect of Disease-Modifying Therapies on Clinical Efficacy of COVID-19 Inactivated Vaccination among People with Multiple Sclerosis. Available at SSRN 3998537. 2022; 73. 73.Schiavetti I, Cordioli C, Stromillo ML, et al. Breakthrough SARS-CoV-2 infections in MS patients on disease modifying therapies. medRxiv. 2022:2022.01.22.22269630. doi:10.1101/2022.01.22.22269630 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMi4wMS4yMi4yMjI2OTYzMHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDIvMTMvMjAyMi4wMi4xMi4yMjI3MDg4My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 74. 74.Liu X, Shaw RH, Stuart ASV, et al. Safety and immunogenicity of heterologous versus homologous prime-boost schedules with an adenoviral vectored and mRNA COVID-19 vaccine (Com-COV): a single-blind, randomised, non-inferiority trial. Lancet. Sep 4 2021;398(10303):856–869. doi:10.1016/s0140-6736(21)01694-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s0140-6736(21)01694-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F02%2F13%2F2022.02.12.22270883.atom) 75. 75.Peiris M, Cheng S, Mok CKP, et al. Neutralizing antibody titres to SARS-CoV-2 Omicron variant and wild-type virus in those with past infection or vaccinated or boosted with mRNA BNT162b2 or inactivated CoronaVac vaccines. Res Sq. Jan 5 2022;doi:10.21203/rs.3.rs-1207071/v1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.21203/rs.3.rs-1207071/v1&link_type=DOI) 76. 76.Kanokudom S, Assawakosri S, Suntronwong N, et al. Safety and Immunogenicity of the Third Booster Dose with Inactivated, Viral Vector, and mRNA COVID-19 Vaccines in Fully Immunized Healthy Adults with Inactivated Vaccine. Vaccines. 2022;10(1):86. 77. 77.Wang X, Zhao X, Song J, et al. Homologous or Heterologous Booster of Inactivated Vaccine Reduces SARS-CoV-2 Omicron Variant Escape from Neutralizing Antibodies. Emerg Microbes Infect. Jan 15 2022:1–18. doi:10.1080/22221751.2022.2030200 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/22221751.2022.2030200&link_type=DOI) 78. 78.Zhang R, Liu D, Leung KY, et al. Immunogenicity of a Heterologous Prime-Boost COVID-19 Vaccination with mRNA and Inactivated Virus Vaccines Compared with Homologous Vaccination Strategy against SARS-CoV-2 Variants. Vaccines (Basel). Jan 3 2022;10(1)doi:10.3390/vaccines10010072 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/vaccines10010072&link_type=DOI) 79. 79.Wanlapakorn N, Suntronwong N, Phowatthanasathian H, et al. Safety and immunogenicity of heterologous and homologous inactivated and adenoviral-vectored COVID-19 vaccines in healthy adults. medRxiv. 2021:2021.11.04.21265908. doi:10.1101/2021.11.04.21265908 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4xMS4wNC4yMTI2NTkwOHYzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDIvMTMvMjAyMi4wMi4xMi4yMjI3MDg4My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 80. 80.Achiron A, Dolev M, Menascu S, et al. COVID-19 vaccination in patients with multiple sclerosis: what we have learnt by February 2021. Multiple Sclerosis Journal. 2021:135245852110034. 81. 81.Achiron A, Mandel M, Dreyer-Alster S, et al. Humoral immune response to COVID-19 mRNA vaccine in patients with multiple sclerosis treated with high-efficacy disease-modifying therapies. Therapeutic advances in neurological disorders. 2021;14:17562864211012835. 82. 82.Gallo A, Capuano R, Donnarumma G, et al. Preliminary evidence of blunted humoral response to SARS-CoV-2 mRNA vaccine in multiple sclerosis patients treated with ocrelizumab. Neurological Sciences. 2021;42(9):3523–3526. 83. 83.van Kempen Z, Wieske L, Stalman E, et al. Longitudinal humoral response after SARS-CoV-2 vaccination in ocrelizumab treated MS patients: To wait and repopulate? Multiple Sclerosis and Related Disorders. 2022;57:103416. 84. 84.Guerrieri S, Lazzarin S, Zanetta C, Nozzolillo A, Filippi M, Moiola L. Serological response to SARS-CoV-2 vaccination in multiple sclerosis patients treated with fingolimod or ocrelizumab: an initial real-life experience. J Neurol. 2021;1:1–5. 85. 85.Novak F, Nilsson AC, Nielsen C, et al. Humoral immune response following SARS-CoV-2 mRNA vaccination concomitant to anti-CD20 therapy in multiple sclerosis. Multiple Sclerosis and Related Disorders. 2021;56:103251.