John Murphy, CEO, The COVID-19 Long-haul Foundation (www.covid19longhaulfoundation.org )
Abstract
COVID‑19 vaccination has altered the trajectory of the pandemic, yet its implications for patients with chronic inflammatory diseases (CIDs) remain nuanced. This review synthesizes evidence across etiology, physiology, genomics, diagnostics, clinical evaluation, therapies, and prognosis. Drawing on peer‑reviewed literature, we examine how vaccines interact with dysregulated immune pathways, the genomic determinants of vaccine response, diagnostic strategies for monitoring immunity, therapeutic adjustments in immunosuppressed patients, and long‑term prognostic considerations.
1. Etiology of Chronic Inflammatory Disease
Chronic inflammatory diseases (CIDs) such as rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, psoriasis, and multiple sclerosis arise from a complex interplay of genetic predisposition, environmental triggers, and immune dysregulation.
- Genetic susceptibility: HLA alleles (e.g., HLA‑DRB1 in RA, HLA‑B27 in spondyloarthropathies) confer risk.
- Environmental factors: Viral infections, smoking, microbiome alterations, and pollutants act as triggers.
- Immune dysregulation: Aberrant activation of innate immunity (TLR signaling, inflammasome activation) and adaptive immunity (autoreactive T and B cells) drive persistent inflammation.
Vaccination context: COVID‑19 vaccines, particularly mRNA platforms, transiently activate innate immunity. In patients with CIDs, this activation may overlap with pre‑existing dysregulated pathways, raising concerns about disease flares. However, large cohort studies suggest that flares are rare and manageable.
2. Physiology of Vaccine Response in CIDs
The physiological response to vaccination involves innate priming and adaptive immunity.
- Innate immunity: mRNA vaccines stimulate toll‑like receptors (TLR7/8), leading to interferon production.
- Adaptive immunity: Antigen presentation activates CD4+ and CD8+ T cells, while B cells generate neutralizing antibodies.
- Cytokine milieu: Patients with CIDs often exhibit elevated IL‑6, TNF‑α, and IFN‑γ, which may alter vaccine efficacy.
Clinical evidence: Immunosuppressed patients (methotrexate, rituximab, corticosteroids) show reduced antibody titers post‑vaccination. Yet, T‑cell responses often remain intact, suggesting partial protection.
3. Genomics and Precision Medicine
Genomic studies reveal that host genetics influence vaccine response.
- HLA associations: Certain alleles predispose to altered immune reactivity.
- Cytokine polymorphisms: IL‑6 and TNF variants modulate inflammatory responses.
- Pharmacogenomics: Genetic markers predict response to biologics (e.g., TNF inhibitors) and may inform vaccine strategies.
Emerging evidence suggests that polygenic risk scores could stratify patients for tailored vaccination schedules, though clinical implementation remains nascent.
4. Diagnostics and Clinical Evaluation
Monitoring vaccine response in CID patients requires serological and cellular assays.
- Serology: Antibody titers (anti‑spike IgG) are often reduced in CID patients.
- Cellular immunity: ELISPOT and flow cytometry reveal preserved T‑cell responses.
- Biomarkers: CRP, ESR, and cytokine panels help track disease activity post‑vaccination.
- Imaging: PET and MRI can detect subclinical inflammation in joints or CNS.
Clinical evaluation should integrate disease activity scores (e.g., DAS28 for RA, SLEDAI for lupus) with immunological monitoring.
5. Therapies and Management
Therapeutic strategies must balance disease control with vaccine efficacy.
- Methotrexate: Temporarily withholding methotrexate around vaccination improves antibody response.
- Biologics: Rituximab impairs humoral immunity; timing vaccination before infusion enhances efficacy.
- Corticosteroids: High doses blunt immune response; tapering may be considered.
- Adjunct therapies: Nutritional support (vitamin D, omega‑3 fatty acids) and microbiome modulation may enhance vaccine response.
Guidelines recommend individualized scheduling of immunosuppressants relative to vaccination.
6. Prognosis
- Short‑term: Most CID patients mount protective responses, though attenuated.
- Long‑term: Vaccination does not worsen CID prognosis; flares are rare and manageable.
- Global perspective: Equitable access remains critical, as CID patients represent a vulnerable population.
Conclusion
COVID‑19 vaccination in chronic inflammatory disease represents a delicate balance between protection and immune modulation. Advances in genomics, diagnostics, and therapeutic strategies promise more precise care. Ongoing research must refine prognostic models and ensure equitable vaccine deployment for vulnerable populations.
1. Etiology of Chronic Inflammatory Disease
1.1 Genetic Predisposition
Chronic inflammatory diseases (CIDs) are fundamentally disorders of immune regulation, and genetics provides the scaffolding upon which environmental triggers act. The human leukocyte antigen (HLA) system is the most consistent genetic determinant. For example, HLA‑DRB104* alleles confer susceptibility to rheumatoid arthritis, while HLA‑B27 is strongly associated with ankylosing spondylitis. These alleles alter antigen presentation, skewing T‑cell repertoires toward autoreactivity.
Beyond HLA, non‑HLA genes such as PTPN22 (a phosphatase regulating T‑cell activation), STAT4 (a transcription factor in cytokine signaling), and TNFAIP3 (a negative regulator of NF‑κB) contribute to disease risk. These polymorphisms collectively lower the threshold for immune activation, creating a milieu in which vaccination‑induced immune stimulation may behave differently than in healthy individuals.
Epigenetic modifications add another layer: DNA methylation patterns in FOXP3 (critical for regulatory T‑cells) and histone acetylation in cytokine promoters have been linked to CID pathogenesis. Such epigenetic landscapes are dynamic, influenced by environment, infection, and therapy, and may modulate vaccine responsiveness.
1.2 Environmental Triggers
Genetic predisposition alone is insufficient; environmental exposures act as catalysts. Viral infections such as Epstein–Barr virus (EBV) have long been implicated in lupus and multiple sclerosis, while gut dysbiosis is central to inflammatory bowel disease.
Lifestyle factors amplify risk. Smoking increases citrullination of proteins, generating neoantigens that drive rheumatoid arthritis. Diets rich in processed foods and low in fiber alter the microbiome, fueling systemic inflammation. Stress and circadian disruption further dysregulate immune homeostasis.
Pollutants — particulate matter, silica, and heavy metals — induce oxidative stress and inflammasome activation, linking environmental toxicity to autoimmunity. These triggers not only initiate disease but also shape how the immune system responds to vaccines, potentially altering cytokine cascades and antibody kinetics.
1.3 Immune Dysregulation
At the core of CIDs lies immune dysregulation.
- Innate immunity: Overactive toll‑like receptors (TLR2, TLR4, TLR7/8) amplify cytokine release. Inflammasomes (NLRP3) generate IL‑1β, perpetuating inflammation.
- Adaptive immunity: Autoreactive CD4+ T cells provide help to B cells, which produce pathogenic autoantibodies (e.g., anti‑CCP in RA, anti‑dsDNA in lupus). CD8+ T cells contribute tissue damage.
- Cytokine networks: IL‑6, TNF‑α, and IFN‑γ orchestrate chronic inflammation, while regulatory cytokines (IL‑10, TGF‑β) are insufficiently expressed.
This dysregulated baseline means that vaccination — which relies on controlled immune activation — occurs against a backdrop of heightened reactivity. Concerns about flares stem from this overlap, though empirical data show most patients tolerate vaccines well.
1.4 Vaccination Context
COVID‑19 vaccines, particularly mRNA platforms, transiently stimulate innate immunity via TLR7/8 and RIG‑I pathways. In healthy individuals, this produces a balanced cytokine surge followed by adaptive immunity. In CIDs, however, the same pathways are already primed. Theoretically, this could exacerbate disease activity.
Large cohort studies, however, demonstrate that flares are rare and generally mild. For example, in rheumatoid arthritis cohorts, fewer than 10% reported transient increases in joint pain post‑vaccination, with no long‑term impact on disease trajectory. In lupus, flares were not significantly different from baseline rates.
Thus, while etiology provides a framework for concern, clinical evidence suggests vaccination is safe and beneficial, even in genetically and immunologically predisposed populations.
2. Physiology of Vaccine Response in CIDs
2.1 Innate Immune Activation
COVID‑19 vaccines initiate immunity through pattern recognition receptors. mRNA vaccines engage TLR7/8, leading to interferon production, while adenoviral vectors stimulate TLR9 and cytosolic sensors. Dendritic cells present antigens, bridging innate and adaptive immunity.
In CIDs, innate immunity is already hyperactive. Elevated baseline interferon signatures in lupus, for example, may alter vaccine kinetics. This raises questions about whether CID patients experience exaggerated or blunted innate responses.
2.2 Adaptive Immunity
Adaptive immunity is the cornerstone of vaccine protection. CD4+ helper T cells orchestrate antibody production, while CD8+ cytotoxic T cells eliminate infected cells. B cells generate neutralizing antibodies against the SARS‑CoV‑2 spike protein.
In CID patients, adaptive immunity is often impaired by therapy. Rituximab depletes B cells, reducing antibody titers. Methotrexate suppresses T‑cell proliferation, blunting cellular immunity. Yet, studies show that T‑cell responses remain relatively preserved, even when antibody titers are reduced. This suggests partial protection persists.
2.3 Altered Physiology in CIDs
Baseline inflammation alters vaccine physiology. Elevated IL‑6 and TNF‑α may interfere with germinal center formation, reducing antibody affinity maturation. Immunosuppressive therapy compounds this effect.
Nevertheless, booster doses restore immunity in many patients. For example, lupus patients on mycophenolate showed improved antibody titers after a third mRNA dose. This highlights the adaptability of the immune system, even in dysregulated states.
2.4 Clinical Implications
- Reduced efficacy: CID patients may require additional booster doses.
- Safety profile: Adverse events are comparable to the general population.
- Monitoring: Disease activity scores (DAS28, SLEDAI) should be tracked post‑vaccination.
3. Genomics and Precision Medicine
3.1 HLA Associations and Vaccine Response
The human leukocyte antigen (HLA) system is central to both autoimmunity and vaccine responsiveness. HLA molecules determine which peptides are presented to T cells, shaping the adaptive immune repertoire.
- Autoimmune predisposition: HLA‑DRB1*04 is linked to rheumatoid arthritis, HLA‑B27 to ankylosing spondylitis, and HLA‑DQ2/DQ8 to celiac disease. These alleles skew antigen presentation toward autoreactive epitopes.
- Vaccine implications: Certain HLA alleles influence the breadth and durability of vaccine‑induced immunity. For example, HLA‑DRB1 variants have been associated with differential antibody titers following influenza vaccination. Emerging data suggest similar variability in COVID‑19 vaccine responses, though large‑scale genomic studies are ongoing.
Thus, HLA typing may eventually guide personalized vaccination schedules in CID patients, identifying those who require booster doses or alternative platforms.
3.2 Cytokine Gene Polymorphisms
Cytokine genes are critical modulators of inflammation and vaccine response.
- IL‑6 polymorphisms: Variants in the IL‑6 promoter region (‑174G/C) influence cytokine expression. High‑expressing alleles correlate with exaggerated inflammatory responses, potentially altering vaccine reactogenicity.
- TNF polymorphisms: TNF‑α promoter variants (‑308G/A) are linked to increased cytokine production, contributing to autoimmunity and possibly heightened vaccine side effects.
- IFN pathway genes: Polymorphisms in IFN regulatory factors (IRF5, IRF7) modulate antiviral responses, influencing both susceptibility to viral infection and vaccine efficacy.
These polymorphisms highlight the genetic heterogeneity underlying vaccine outcomes in CID patients.
3.3 Pharmacogenomics and Biologic Therapies
Pharmacogenomics explores how genetic variation influences drug response. In CIDs, biologic therapies such as TNF inhibitors, IL‑6 blockers, and B‑cell depleting agents are widely used.
- TNF inhibitors: Genetic markers in TNF and Fc receptor genes predict therapeutic response. Patients with poor biologic response may also exhibit altered vaccine immunogenicity.
- Rituximab: B‑cell depletion impairs humoral immunity. Genetic variants in CD20 and FcγR genes influence both drug efficacy and vaccine antibody production.
- JAK inhibitors: Polymorphisms in JAK/STAT pathway genes modulate drug response and may affect vaccine‑induced cytokine signaling.
Pharmacogenomic profiling could inform both therapy selection and vaccination timing, optimizing outcomes in CID patients.
3.4 Polygenic Risk Scores and Predictive Models
Polygenic risk scores (PRS) integrate multiple genetic variants to estimate disease risk. In autoimmunity, PRS models have been developed for rheumatoid arthritis, lupus, and IBD.
- Predicting vaccine response: PRS could stratify patients into high‑risk groups for poor vaccine immunogenicity.
- Clinical utility: While not yet routine, PRS may guide personalized vaccination strategies, identifying patients who require enhanced monitoring or alternative platforms.
- Future directions: Integration of PRS with transcriptomic and proteomic data could yield predictive models of vaccine efficacy and safety in CID populations.
3.5 Ethical and Equity Considerations
Genomic medicine raises ethical questions. Access to HLA typing and pharmacogenomic testing is uneven globally. Precision vaccination strategies must avoid exacerbating health inequities. Moreover, genomic data privacy is paramount, particularly when linked to sensitive health conditions.
3.6 Clinical Implications
- Personalized vaccination: Genomic profiling may identify patients who benefit from tailored schedules.
- Risk stratification: Genetic markers could predict adverse events or poor immunogenicity.
- Integration with therapy: Pharmacogenomics may guide timing of immunosuppressants relative to vaccination.
4. Diagnostics and Clinical Evaluation
4.1 Serological Assays
Serological testing remains the most accessible method for evaluating vaccine response. Measurement of anti‑spike IgG titers provides a quantitative assessment of humoral immunity. In healthy populations, robust antibody responses are observed after two doses of mRNA vaccines. However, in patients with chronic inflammatory diseases (CIDs), titers are often reduced.
- Rheumatoid arthritis: Patients on methotrexate exhibit diminished antibody levels, though temporary drug withdrawal improves titers.
- Systemic lupus erythematosus (SLE): Antibody responses are variable, with some patients showing delayed seroconversion.
- Inflammatory bowel disease (IBD): Anti‑TNF therapy reduces antibody titers, while vedolizumab appears less impactful.
Serology is limited by its inability to capture cellular immunity, yet it remains a cornerstone of clinical evaluation.
4.2 Cellular Immunity Testing
Cellular immunity provides a deeper understanding of vaccine protection. Techniques such as ELISPOT assays, intracellular cytokine staining, and flow cytometry measure T‑cell reactivity to SARS‑CoV‑2 antigens.
- CD4+ T cells: Critical for orchestrating antibody production.
- CD8+ T cells: Provide cytotoxic defense against infected cells.
- Memory T cells: Ensure long‑term protection.
Studies show that even when antibody titers are reduced, CID patients often retain robust T‑cell responses. This underscores the importance of cellular assays in complementing serology.
4.3 Biomarkers of Inflammation
Monitoring disease activity post‑vaccination requires biomarkers.
- C‑reactive protein (CRP) and erythrocyte sedimentation rate (ESR) are traditional markers of systemic inflammation.
- Cytokine panels (IL‑6, TNF‑α, IFN‑γ) provide more granular insights.
- Autoantibody levels (anti‑dsDNA, anti‑CCP) may fluctuate post‑vaccination, though clinical significance is limited.
Biomarker monitoring helps distinguish vaccine‑related immune activation from disease flares.
4.4 Imaging Modalities
Imaging offers non‑invasive evaluation of inflammation.
- PET scans: Detect metabolic activity in inflamed tissues.
- MRI: Useful for CNS inflammation in multiple sclerosis.
- Ultrasound: Assesses synovitis in rheumatoid arthritis.
Post‑vaccination imaging studies show no evidence of sustained inflammation, supporting vaccine safety in CID patients.
4.5 Disease Activity Indices
Clinical evaluation integrates laboratory and imaging data with standardized indices.
- DAS28: Disease Activity Score for rheumatoid arthritis.
- SLEDAI: Systemic Lupus Erythematosus Disease Activity Index.
- Mayo score: For ulcerative colitis.
- EDSS: Expanded Disability Status Scale for multiple sclerosis.
Tracking these indices post‑vaccination ensures that transient symptoms are not misinterpreted as disease progression.
4.6 Integrated Diagnostic Approach
Optimal evaluation combines serology, cellular assays, biomarkers, imaging, and clinical indices. This holistic approach distinguishes vaccine‑related immune activation from true disease flares, guiding therapeutic decisions.
5. Therapies and Management
5.1 Conventional Immunosuppressants
Methotrexate remains the cornerstone of therapy in rheumatoid arthritis and other autoimmune conditions. However, methotrexate impairs vaccine immunogenicity by suppressing lymphocyte proliferation. Clinical trials demonstrate that temporary discontinuation of methotrexate for 1–2 weeks around COVID‑19 vaccination significantly improves antibody titers without causing major disease flares. Other conventional agents such as azathioprine and mycophenolate mofetil similarly reduce vaccine responses, though the magnitude varies. Balancing disease control with vaccine efficacy requires individualized strategies.
5.2 Biologic Therapies
Biologics target specific cytokines or immune cells, profoundly altering vaccine responses.
- TNF inhibitors (infliximab, adalimumab): Reduce antibody titers but generally preserve T‑cell immunity.
- IL‑6 inhibitors (tocilizumab): May blunt acute vaccine reactogenicity but do not significantly impair immunogenicity.
- B‑cell depleting agents (rituximab): Profoundly impair humoral immunity, often eliminating antibody responses. Timing vaccination before rituximab infusion is critical.
- JAK inhibitors (tofacitinib, baricitinib): Modulate cytokine signaling; data suggest modest reductions in vaccine efficacy.
Biologics necessitate careful scheduling of vaccination relative to therapy cycles.
5.3 Corticosteroid Modulation
Corticosteroids are widely used for acute disease control but are immunosuppressive. High‑dose corticosteroids (>20 mg/day prednisone equivalent) blunt both humoral and cellular vaccine responses. Strategies include tapering corticosteroids before vaccination when clinically feasible, though disease stability must be prioritized. Low‑dose corticosteroids appear less impactful.
5.4 Timing Strategies Around Vaccination
Optimal vaccine efficacy in CID patients often requires strategic timing of immunosuppressants.
- Methotrexate: Withhold for 1–2 weeks post‑vaccination.
- Rituximab: Vaccinate at least 4 weeks before infusion or 6 months after last dose.
- TNF inhibitors: No adjustment typically required, though monitoring is advised.
- JAK inhibitors: Consider brief interruption if disease control allows.
These strategies are supported by consensus guidelines from rheumatology and immunology societies.
5.5 Adjunctive Nutritional and Lifestyle Interventions
Adjunctive measures may enhance vaccine response.
- Vitamin D: Supports immune regulation; deficiency is linked to poor vaccine outcomes.
- Omega‑3 fatty acids: Anti‑inflammatory properties may improve immune balance.
- Microbiome modulation: Probiotics and dietary fiber support gut immunity, potentially enhancing vaccine efficacy.
- Exercise and stress reduction: Improve immune resilience and may reduce flare risk.
While evidence is preliminary, these interventions represent low‑risk strategies to optimize outcomes.
5.6 Clinical Guidelines and Consensus
Professional societies (EULAR, ACR, BSG) recommend vaccination for all CID patients, with adjustments based on therapy. The consensus emphasizes that the benefits of vaccination outweigh theoretical risks of disease flare, and that therapy modification should be individualized.
5.7 Integrated Management Approach
Effective management requires collaboration between rheumatologists, gastroenterologists, dermatologists, and immunologists. Shared decision‑making with patients is essential, balancing disease control with vaccine protection.
6. Prognosis
6.1 Short‑Term Outcomes
In the immediate aftermath of COVID‑19 vaccination, most patients with chronic inflammatory diseases (CIDs) mount protective immune responses.
- Humoral immunity: Antibody titers are often lower than in healthy controls, particularly in patients receiving methotrexate, rituximab, or high‑dose corticosteroids. However, even attenuated titers confer meaningful protection against severe disease.
- Cellular immunity: T‑cell responses remain relatively intact across CID populations, ensuring a baseline of protection even when antibodies are reduced.
- Disease flares: Transient increases in symptoms (arthralgia, fatigue, rash) occur in a minority of patients, but large cohort studies show flare rates are not significantly higher than baseline.
Thus, short‑term prognosis following vaccination is favorable, with protection outweighing risks.
6.2 Long‑Term Disease Trajectory
Long‑term outcomes reflect both vaccine durability and CID progression.
- Durability of immunity: Antibody waning occurs more rapidly in immunosuppressed patients, necessitating booster doses.
- Disease progression: Vaccination does not accelerate CID progression. Longitudinal studies in RA, lupus, and IBD show stable disease activity over 12–18 months post‑vaccination.
- Breakthrough infections: While more common in CID patients, breakthrough infections are generally mild, underscoring the protective effect of vaccination.
Overall, vaccination does not worsen long‑term prognosis and may reduce morbidity by preventing severe COVID‑19.
6.3 Global Equity and Access
Prognosis is shaped not only by biology but also by access.
- High‑income countries: CID patients benefit from booster programs, genomic monitoring, and tailored therapy adjustments.
- Low‑ and middle‑income countries: Limited access to vaccines and biologics exacerbates vulnerability.
- Equity imperative: Global health organizations emphasize equitable distribution, recognizing CID patients as a priority group.
Without equitable access, prognosis diverges sharply across regions, highlighting the ethical dimension of vaccination.
6.4 Future Directions in Personalized Vaccination
The future of prognosis lies in precision medicine.
- Genomic stratification: HLA typing and cytokine polymorphisms may predict vaccine response.
- Pharmacogenomics: Integration of drug response profiles with vaccination schedules will optimize outcomes.
- Systems biology: Multi‑omics approaches (genomics, transcriptomics, proteomics) will yield predictive models of vaccine efficacy.
- Digital health: Wearable devices and AI‑driven monitoring may track disease activity and vaccine response in real time.
These innovations promise a future where prognosis is not generalized but personalized, ensuring optimal protection for each patient.
6.5 Integrated Prognostic Model
Prognosis in CID patients post‑vaccination is best understood as a multifactorial model:
- Biological factors: Genetics, immune dysregulation, therapy.
- Clinical factors: Disease activity, comorbidities.
- Social factors: Access, equity, patient education.
Such integrated models will guide clinicians in tailoring vaccination strategies, improving both individual and population outcomes.
References
- Smolen JS, et al. Nat Rev Immunol. 2016.
- Tsokos GC. N Engl J Med. 2011.
- Belkaid Y, Hand TW. Science. 2014.
- Kaplan GG. Gastroenterology. 2011.
- Furer V, et al. Ann Rheum Dis. 2021.
- Kawai T, Akira S. Nat Immunol. 2010.
- Boyarsky BJ, et al. JAMA. 2021.
- Deepak P, et al. Gastroenterology. 2021.
- McInnes IB, Schett G. Nat Rev Immunol. 2007.
- Raychaudhuri S, et al. Nature. 2012.
- Fishman D, et al. Genes Immun. 1998.
- Weinshilboum R, Wang L. Nat Rev Drug Discov. 2017.
- Simon D, et al. Arthritis Rheum. 2021.
- Sattler A, et al. Nat Med. 2021.
- van der Heijde D, et al. Ann Rheum Dis. 2017.
- Park JK, et al. Arthritis Care Res. 2019.
- Curtis JR, et al. Arthritis Rheum. 2016.
- Furer V, et al. Ann Rheum Dis. 2021.
- Calder PC. Nutrients. 2020.
- Mahil SK, et al. Lancet. 2021.
- Boekel L, et al. Ann Rheum Dis. 2022.
- Braun-Moscovici Y, et al. Arthritis Rheum. 2021.
- WHO. Lancet. 2021.
- Barouch DH. Nat Rev Immunol. 2022.
- Polack FP, et al. N Engl J Med. 2020.
- Raychaudhuri S, et al. Nature. 2012.
- Fishman D, et al. Genes Immun. 1998.
- McInnes IB, Schett G. Nat Rev Immunol. 2007.
- Weinshilboum R, Wang L. Nat Rev Drug Discov. 2017.
- Furer V, et al. Ann Rheum Dis. 2021.
- Boyarsky BJ, et al. JAMA. 2021.
- Deepak P, et al. Gastroenterology. 2021.
- Sattler A, et al. Nat Med. 2021.
- Mahil SK, et al. Lancet. 2021.
- Simon D, et al. Antibody response in RA patients. Arthritis Rheum. 2021.
- Deepak P, et al. COVID‑19 vaccine immunogenicity in IBD. Gastroenterology. 2021.
- Sattler A, et al. T‑cell immunity after vaccination. Nat Med. 2021.
- Braun‑Moscovici Y, et al. Vaccine safety in RA. Arthritis Rheum. 2021.
- van der Heijde D, et al. Imaging in inflammatory arthritis. Ann Rheum Dis. 2017.
- Mahil SK, et al. Vaccine response in psoriasis. Lancet. 2021.
- Boekel L, et al. Long‑term outcomes post‑vaccination in autoimmune disease. Ann Rheum Dis. 2022.
- Park JK, et al. Methotrexate and vaccine response. Arthritis Care Res. 2019.
- Curtis JR, et al. Biologics and immunogenicity. Arthritis Rheum. 2016.
- Furer V, et al. Timing immunosuppressants with vaccination. Ann Rheum Dis. 2021.
- Deepak P, et al. Vaccine immunogenicity in IBD. Gastroenterology. 2021.
- Braun‑Moscovici Y, et al. Vaccine safety in RA. Arthritis Rheum. 2021.
- Calder PC. Nutrition and immunity. Nutrients. 2020.
- Mahil SK, et al. Vaccine response in psoriasis. Lancet. 2021.
- Boekel L, et al. Long‑term outcomes post‑vaccination in autoimmune disease. Ann Rheum Dis. 2022.
- Braun‑Moscovici Y, et al. Vaccine safety in RA. Arthritis Rheum. 2021.
- Boyarsky BJ, et al. Vaccine response in immunosuppressed patients. JAMA. 2021.
- Deepak P, et al. COVID‑19 vaccine immunogenicity in IBD. Gastroenterology. 2021.
- WHO. Global vaccine equity. Lancet. 2021.
- Barouch DH. COVID‑19 vaccines: global perspective. Nat Rev Immunol. 2022.