1
|
Mahroum N, Elsalti A, Al Shawaf M, Darkhabani M, Alwani A, Seida R, Ertas MT, Simsek AG, Awad M, Habra M, Alrifaai MA, Bogdanos D, Shoenfeld Y. Artificial intelligence meets the world experts; updates and novel therapies in autoimmunity - The 14th international congress on autoimmunity 2024 (AUTO14), Ljubljana. Autoimmun Rev 2025; 24:103698. [PMID: 39571671 DOI: 10.1016/j.autrev.2024.103698] [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: 06/21/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
The bi-annual international congress on autoimmunity is a huge opportunity for the medical community to discuss the latest updates in the field. During the 14th congress 2024 (AUTO14) in Ljubljana, artificial intelligence (AI) occupied special attention due to its recent and ongoing unequivocal role in various medical fields including autoimmunity. For instance, through a challenging debate between world-experts and the most popular AI bot used (ChatGPT), several clinical cases including a case of vasculitis were discussed in the plenary sessions. ChatGPT agreed with the clinical decisions made by the experts nevertheless, the bot added additional aspects related to the specific case. In this regard, ChatGPT emphasized the need for osteoporosis prophylaxis in a patient planned to be treated with systemic steroids for a long time. Furthermore, AUTO14 included the newest updates on most autoimmune disorders, distributed among tens of sessions. Among others, infection and autoimmunity, the sequalae of the pandemic of COVID-19, as well as COVID-19 vaccines were discussed as well. Due to the high numbers of the works presented, and for ensuring that important updates are not missed; we divided our paper into sections. The subtitles throughout the paper correspond to different sessions of the congress, all presenting new updates in the field. A figure aiding in navigating throughout the paper was also provided.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Maisam Al Shawaf
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mohammad Darkhabani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulrahman Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | | | - Mustafa Awad
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Dimitrios Bogdanos
- Department of Rheumatology and Clinical Immunology, University Hospital of Larisa, School of Medicine, University of Thessaly, Larissa, Greece
| | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
2
|
Rahman M. Editorial: design considerations for future personalized vaccination approaches. Nanomedicine (Lond) 2024:1-6. [PMID: 39552583 DOI: 10.1080/17435889.2024.2419816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Affiliation(s)
- Mahbuba Rahman
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
3
|
Rodríguez-García FA, Servín-Rodríguez CE, Torres-Salazar QL. Autoimmune/inflammatory syndrome induced by adjuvants (ASIA): A case of systemic symptoms following breast implants and vaccinations. Int J Surg Case Rep 2024; 124:110497. [PMID: 39442271 PMCID: PMC11539708 DOI: 10.1016/j.ijscr.2024.110497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION AND IMPORTANCE The Autoimmune/inflammatory Syndrome Induced by Adjuvants (ASIA) is a recently recognized entity characterized by a constellation of nonspecific symptoms that develop after exposure to adjuvants. Adjuvants can include vaccines, silicone, and other foreign substances. Here, we present the case of a 31-year-old woman who developed ASIA syndrome following breast implants and booster vaccinations, emphasizing the diagnostic challenges and treatment considerations. CASE PRESENTATION A 31-year-old female presented with a history of progressive systemic symptoms including chronic fatigue, myalgia, arthralgia, and headaches, four months after receiving breast implants. Her clinical history was further complicated by multiple vaccinations, including influenza and SARS-CoV-2 boosters. After extensive diagnostic workup and unsuccessful medical management, she was diagnosed with ASIA syndrome. The decision was made to remove the implants, leading to rapid and complete resolution of her symptoms. CLINICAL DISCUSSION ASIA syndrome is a difficult diagnosis due to its broad symptomatology and its mimicry of other autoimmune and inflammatory conditions. Diagnostic criteria, proposed by Shoenfeld et al., include exposure to adjuvants and the appearance of typical clinical manifestations such as fatigue, myalgias, and arthralgias. In this case, the temporal association with breast implants and vaccinations made the diagnosis more evident. Surgical removal of the implants led to immediate improvement, reinforcing the diagnosis. CONCLUSION This case highlights the importance of recognizing ASIA syndrome as a potential diagnosis in patients with exposure to adjuvants and unexplained systemic symptoms. Early diagnosis and intervention, such as removal of the triggering adjuvant, are essential for patient recovery. EVIDENCE BASED MEDICINE RANKING Level IV.
Collapse
|
4
|
Mitsuyama H, Iizasa E, Kukita A, Toda S, Yoshida H, Inoue H, Hara H. Deletion of Card9 eliminates the detrimental facets of mycobacterial adjuvants. Heliyon 2024; 10:e38139. [PMID: 39386804 PMCID: PMC11462255 DOI: 10.1016/j.heliyon.2024.e38139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Although mycobacterial adjuvants are capable of eliciting a strong adaptive humoral and cellular immunity, they also sometimes provoke detrimental outcomes, including autoimmune/inflammatory syndromes. Here, we show that the deletion of caspase recruitment domain family member 9 (Card9), a signaling adaptor of a set of innate immune receptors, can eliminate the detrimental effects of mycobacterial adjuvants. Long-lasting tissue-destructive skin inflammation at the site of complete Freund's adjuvant (CFA) injection, lung granuloma formation induced by intratracheal Mycobacterium bovis Bacillus Calmette-Guérin infection, and the incidence and severity of experimental autoimmune encephalomyelitis and collagen-induced arthritis induced by autoantigen immunization with CFA were considerably attenuated in Card9-deficient (Card9 -/- ) mice compared to control wild-type mice. Card9 -/- mice showed impaired development of Th17, but not Th1, in the early phase after autoimmune induction, due to the impaired development of IL-6-producing Sirpαhigh dendritic cells, which are essential for priming pathigenic Th17, in the draining lymph nodes. However, Card9 deletion did not affect overall adaptive antibody production or delayed-type hypersensitivity following immunization with CFA, indicating that humoral and type 1 immune responses remained intact. These results suggest that avoiding the activation of Card9 signaling during vaccination with mycobacteria-containing vaccines may mitigate the risk of detrimental type 3 immune responses, while preserving type 1 immune responses that are effective against intracellular pathogens and cancers.
Collapse
Affiliation(s)
- Hideo Mitsuyama
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Ei'ichi Iizasa
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
- Department of Psychosomatic Internal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Akiko Kukita
- Research Center of Arthroplasty, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga. 849-8501, Japan
| | - Shuji Toda
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga, 849-8501, Japan
- Department of Pathology, Takagi Hospital, Okawa, Fukuoka, 831-8501, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga, 849-8501, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| |
Collapse
|
5
|
Yildiz Tasci Y, Icoz M, Gurturk Icoz SG, Saritas O, Arikan Yorgun M, Toklu Y. Evaluation of the early effects of the first-dose administration of the Sinovac vaccine on the retina, choroid, and optic disc using optical coherence tomography (OCT) and OCT-angiography. Cutan Ocul Toxicol 2024:1-7. [PMID: 39383017 DOI: 10.1080/15569527.2024.2408683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024]
Abstract
AIM To determine the effects of the first-dose administration of the Sinovac vaccine on the retina, choroid, and optic disc in healthy participants. METHODS This prospective design study was conducted with 27 healthy healthcare workers who received the first dose of Sinovac vaccine and 25 healthy controls who were not vaccinated. In the vaccinated group, ophthalmological examinations and measurements were performed before vaccination and one week and one month after vaccination. Subfoveal, nasal, and temporal choroidal thicknesses (CTs), retinal nerve fiber layer (RNFL) thickness, and macular thickness (MT) were determined using spectral domain-optical coherence tomography at all visits. Superficial, deep, and peripapillary radial capillary plexus (superficial capillary plexus, deep capillary plexus (DCP), and radial peripapillary capillary, respectively), choriocapillaris vascular density, and foveal avascular zone parameters were measured on optical coherence tomography-angiography (OCT-A). RESULTS No significant difference was detected between the two groups in terms of the parameters measured by OCT and OCT-A (p > 0.05 for all). The CT values measured in all quadrants were significantly higher at the first week after vaccination (p < 0.05 for all), and they returned to their pre-vaccination values at the first month post-vaccination measurement (p > 0.05 for all). Concerning the RNFL and MT values, there was no significant difference between the pre-vaccination and post-vaccination first-week measurements (p > 0.05 for all), but a statistically significant increase was detected in the post-vaccination first-month MT and RNFL measurements (p < 0.05 for all). Only the decreases in the foveal DCP and choriocapillaris vascular density values were significant at the first week after vaccination (p < 0.05 for all). CONCLUSION The early changes detected after vaccination in this study suggest the possibility that autoimmune, vascular, and inflammatory diseases may simultaneously emerge in the early post-vaccination period or may be triggered after vaccination, or that the vaccine may unmask these diseases.
Collapse
Affiliation(s)
- Yelda Yildiz Tasci
- Department of Ophthalmology, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| | - Mehmet Icoz
- Department of Ophthalmology, Yozgat City Hospital, Yozgat, Turkey
| | | | - Ozge Saritas
- Department of Ophthalmology, Battalgazi State Hospital, Malatya, Turkey
| | - Mucella Arikan Yorgun
- Department of Ophthalmology, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| | - Yasin Toklu
- Department of Ophthalmology, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
6
|
David P, Santos GDM, Patt YS, Orsi FA, Shoenfeld Y. Immune thrombocytopenia (ITP) - could it be part of autoimmune/inflammatory syndrome induced by adjuvants (ASIA)? Autoimmun Rev 2024; 23:103605. [PMID: 39182594 DOI: 10.1016/j.autrev.2024.103605] [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: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 08/27/2024]
Abstract
Immune thrombocytopenia (ITP) is a complex autoimmune disorder characterized by thrombocytopenia and an increased bleeding risk, arising from autoantibody-mediated platelet destruction and impaired megakaryocyte function. The pathogenesis of ITP involves a multifaceted interplay of genetic predispositions, immune dysregulation, and environmental triggers, though the precise mechanisms remain uncertain. Several infectious agents, mostly viruses, have been implicated in both acute and chronic ITP through mechanisms such as molecular mimicry, direct bone marrow suppression, and immune dysregulation. Vaccinations, particularly those containing adjuvants like aluminum and those capable of inducing molecular mimicry, have also been associated with ITP, either as a new onset or as a relapse in preexisting cases. The role of drugs, particularly quinine, quinidine and certain antibiotics, in inducing ITP through various immunological pathways further illustrates the diverse etiologies of this condition. The multiple triggers of the disease raise the question of whether ITP may be classified as an autoimmune/inflammatory syndrome induced by adjuvants (ASIA). This condition encompasses a range of autoimmune and inflammatory symptoms triggered by adjuvants, such as silicones, polypropylene meshes, metal implants, and mineral oils present in various medical materials and medications. Similar to that observed in some cases of ITP, adjuvants can trigger autoimmune or autoinflammatory responses via molecular mimicry, epitope spreading, and polyclonal activation. This narrative review explores the underlying environmental factors related to ITP and examines ITP triggers that could potentially support an association between ITP and ASIA syndrome.
Collapse
Affiliation(s)
- Paula David
- Internal Medicine B, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Leeds Institute of Rheumatic and Musculoskeletal Diseases (LIRMM), University of Leeds, Leeds, UK.
| | - Gabrielle de Mello Santos
- Hospital das Clinicas of University of São Paulo Medical School (HCFMUSP), São Paulo, Brazil; HEMORIO - State Institute of Hematology "Arthur de Siqueira Cavalcanti", Rio de Janeiro, Brazil
| | - Yonatan Shneor Patt
- Internal Medicine B, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Fernanda A Orsi
- Hospital das Clinicas of University of São Paulo Medical School (HCFMUSP), São Paulo, Brazil; Department of Pathology, School of Medical Sciences of the University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Yehuda Shoenfeld
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
7
|
Granja López J, Estebas Armas C, Lorenzo Dieguez M, Puertas Muñoz I, De Celis Ruiz E, Rigual R, Fernández-Fournier M, Torres Iglesias G, Sánchez Velasco S, Tallón Barranco A, Rogozina O, Ramírez E, González-Muñoz M, Lacruz Ballester L. Neurological manifestations of immune origin after COVID-19 vaccination: retrospective case study. Front Pharmacol 2024; 15:1376474. [PMID: 39175548 PMCID: PMC11338880 DOI: 10.3389/fphar.2024.1376474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024] Open
Abstract
Objectives: To know the frequency and characteristics of neurological manifestations of probable immune origin occurring after exposure to COVID-19 vaccination. In addition, to pre-study the usefulness of the Spanish pharmacovigilance system and lymphocyte transformation test in establishing causality. Methods: Retrospective case study, including patients admitted to the Neurology department from January 2021 to May 2022 with a probable neuroimmune disorder. Demographic, clinical and COVID-19 vaccination antecedent data were collected from medical records. Results: From a total of 108 patients, 30 were excluded due to a different etiological diagnosis after follow-up. Thirty-six patients (46.2%) had received the COVID-19 vaccine in the previous 3 months (21.8% during the previous month). BioNTech-Pfizer vaccine was the most frequent in this group (63.9%). 69/108 were female and mean age 51.2 years (SD 22.59), with no significant difference with not recently-vaccinated (U-Mann Whitney, p = 0.256). The neurological syndromes found were (vaccinated/total): polyradiculoneuropathy (8/16), encephalitis (5/11), multiple sclerosis relapse (5/16), optic neuritis (1/4), myelitis (3/6), cranial neuropathy (6/10), aseptic meningitis (1/3) and others (7/11). Acute immunosuppressive treatment was administered in 61.1% of cases and 47.2% presented complete clinical improvement, without significant differences with non-vaccinated patients (chi-square, p = 0.570). Eleven vaccinated patients were studied in the pharmacovigilance office for possible adverse drug reaction. Causality according to the Spanish pharmacovigilance system (SPVS) algorithm was "Related" to COVID-19 vaccine (score ≥ 4) in 11 cases with positive in vitro study (lymphocyte transformation test) to polyethylene glycol-2000 and polysorbate-80 in 4 cases. Conclusion: Neuroimmune disorders appearing after administration of COVID-19 vaccine do not seem to present important differentiating clinical and/or evolutive features. Delayed hypersensitivity to vaccine excipients could be one of the pathophysiological mechanisms, and lymphocyte transformation test is a useful tool to identify it.
Collapse
Affiliation(s)
- Juan Granja López
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Carlos Estebas Armas
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Manuel Lorenzo Dieguez
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Inmaculada Puertas Muñoz
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Elena De Celis Ruiz
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Ricardo Rigual
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Mireya Fernández-Fournier
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Gabriel Torres Iglesias
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Sara Sánchez Velasco
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Antonio Tallón Barranco
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| | - Olga Rogozina
- Clinical Pharmacology Department, La Paz University Hospital-IdiPAZ, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Ramírez
- Clinical Pharmacology Department, La Paz University Hospital-IdiPAZ, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Laura Lacruz Ballester
- Neurology Department and Stroke Center, Laboratory of Neurological and Cerebrovascular Sciences, Hospital La Paz Institute for Health Research Institute-IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), Madrid, Spain
| |
Collapse
|
8
|
Bălăceanu-Gurău B, Dumitrascu A, Giurcăneanu C, Tatar R, Gurău CD, Orzan OA. A Comprehensive Review on the Intricate Interplay between COVID-19 Immunization and the New Onset of Pemphigus Foliaceus. Vaccines (Basel) 2024; 12:857. [PMID: 39203983 PMCID: PMC11360219 DOI: 10.3390/vaccines12080857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Autoimmune bullous diseases (AIBDs) are characterized by the formation of vesicles, bullous lesions, and mucosal erosions. The autoantibodies target the cellular anchoring structures from the surface of epidermal keratinocyte named desmosomes, leading to a loss of cellular cohesion named acantholysis. AIBDs are classified into intraepidermal or subepidermal types based on clinical features, histological characteristics, and immunofluorescence patterns. Pemphigus foliaceus (PF) is an acquired, rare, autoimmune skin condition associated with autoantibodies that specifically target desmoglein-1, leading to a clinical presentation characterized by delicate cutaneous blisters, typically sparing the mucous membranes. Several factors, including genetic predisposition, environmental triggers, malignancies, medication use, and vaccination (for influenza, hepatitis B, rabies, tetanus, and more recently, severe acute respiratory syndrome Coronavirus 2 known as SARS-CoV-2), can potentially trigger the onset of pemphigus. With the advent of vaccines playing a pivotal role in combatting the 2019 coronavirus disease (COVID-19), extensive research has been conducted globally to ascertain their efficacy and potential cutaneous adverse effects. While reports of AIBDs post-COVID-19 vaccination exist in the medical literature, instances of PF following vaccination have been less commonly reported worldwide. The disease's pathophysiology is likely attributed to the resemblance between the ribonucleic acid (RNA) antigen present in these vaccines and cellular nuclear matter. The protein produced by the BNT-162b2 messenger ribonucleic acid (mRNA) vaccine includes immunogenic epitopes that could potentially trigger autoimmune phenomena in predisposed individuals through several mechanisms, including molecular mimicry, the activation of pattern recognition receptors, the polyclonal stimulation of B cells, type I interferon production, and autoinflammation. In this review, we present a comprehensive examination of the existing literature regarding the relationship between COVID-19 and PF, delving into their intricate interactions. This exploration improves the understanding of both pemphigus and mRNA vaccine mechanisms, highlighting the importance of close monitoring for PF post-immunization.
Collapse
Affiliation(s)
- Beatrice Bălăceanu-Gurău
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (O.A.O.)
- Clinic of Dermatology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| | - Adrian Dumitrascu
- Division of Hospital Internal Medicine, Department of Medicine, Mayo Clinic Florida, Jacksonville, FL 32224, USA;
| | - Călin Giurcăneanu
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (O.A.O.)
- Clinic of Dermatology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| | - Raluca Tatar
- Department of Plastic Reconstructive Surgery and Burns, “Grigore Alexandrescu” Clinical Emergency Hospital for Children, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Plastic Reconstructive Surgery and Burns, “Grigore Alexandrescu” Clinical Emergency Hospital for Children, 010621 Bucharest, Romania
| | - Cristian-Dorin Gurău
- Orthopedics and Traumatology Clinic, Clinical Emergency Hospital, 014451 Bucharest, Romania;
| | - Olguța Anca Orzan
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (O.A.O.)
- Clinic of Dermatology, “Elias” Emergency University Hospital, 011461 Bucharest, Romania
| |
Collapse
|
9
|
Ganhör C, Mayr L, Zolles J, Almeder M, Kazemi M, Mandl M, Wechselberger C, Bandke D, Theiner S, Doppler C, Schweikert A, Müller M, Puh Š, Kotnik M, Langer R, Koellensperger G, Bernhard D. Airborne Aluminum as an Underestimated Source of Human Exposure: Quantification of Aluminum in 24 Human Tissue Types Reveals High Aluminum Concentrations in Lung and Hilar Lymph Node Tissues. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:11292-11300. [PMID: 38888518 PMCID: PMC11223461 DOI: 10.1021/acs.est.4c01910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Aluminum (Al) is the most abundant metal in the earth's crust, and humans are exposed to Al through sources like food, cosmetics, and medication. So far, no comprehensive data on the Al distribution between and within human tissues were reported. We measured Al concentrations in 24 different tissue types of 8 autopsied patients using ICP-MS/MS (inductively coupled plasma-tandem mass spectrometry) under cleanroom conditions and found surprisingly high concentrations in both the upper and inferior lobes of the lung and hilar lymph nodes. Al/Si ratios in lung and hilar lymph node samples of 12 additional patients were similar to the ratios reported in urban fine dust. Histological analyses using lumogallion staining showed Al in lung erythrocytes and macrophages, indicating the uptake of airborne Al in the bloodstream. Furthermore, Al was continuously found in PM2.5 and PM10 fine dust particles over 7 years in Upper Austria, Austria. According to our findings, air pollution needs to be reconsidered as a major Al source for humans and the environment.
Collapse
Affiliation(s)
- Clara Ganhör
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Lukas Mayr
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Julia Zolles
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Marion Almeder
- Institute
of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz 4020, Austria
| | - Matin Kazemi
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Markus Mandl
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Christian Wechselberger
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Dave Bandke
- Institute
of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz 4020, Austria
| | - Sarah Theiner
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Christian Doppler
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Andreas Schweikert
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Marina Müller
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Špela Puh
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Michaela Kotnik
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| | - Rupert Langer
- Institute
of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz 4020, Austria
| | - Gunda Koellensperger
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - David Bernhard
- Division
of Pathophysiology, Institute of Physiology and Pathophysiology, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
- Clinical
Research Institute for Cardiovascular and Metabolic Diseases, Medical
Faculty, Johannes Kepler University, Linz 4020, Austria
| |
Collapse
|
10
|
Chen Y, Song X, Chen W, Zhao X, Yang L, Liu D. Epitope screening and self-assembled nanovaccine molecule design of PDCoV-S protein based on immunoinformatics. Front Microbiol 2024; 15:1402963. [PMID: 38903798 PMCID: PMC11186991 DOI: 10.3389/fmicb.2024.1402963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Based on the whole virus or spike protein of pigs, δ coronavirus (PDCoV) as an immunogen may have unrelated antigenic epitope interference. Therefore, it is essential for screening and identifying advantageous protective antigen epitopes. In addition, immunoinformatic tools are described as an important aid in determining protective antigenic epitopes. In this study, the primary, secondary, and tertiary structures of vaccines were measured using ExPASy, PSIPRED 4.0, and trRosetta servers. Meanwhile, the molecular docking analysis and vector of the candidate nanovaccine were constructed. The immune response of the candidate vaccine was simulated and predicted using the C-ImmSim server. This experiment screened B cell epitopes with strong immunogenicity and high conservation, CTL epitopes, and Th epitopes with IFN-γ and IL-4 positive spike proteins. Ferritin is used as a self-assembled nanoparticle element for designing candidate nanovaccine. After analysis, it has been found to be soluble, stable, non-allergenic, and has a high affinity for its target receptor, TLR-3. The preliminary simulation analysis results show that the candidate nanovaccine has the ability to induce a humoral and cellular immune response. Therefore, it may provide a new theoretical basis for research on coronavirus self-assembled nanovaccines. It may be an effective candidate vaccine for controlling and preventing PDCoV.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongyu Liu
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| |
Collapse
|
11
|
Anderson E, Powell M, Yang E, Kar A, Leung TM, Sison C, Steinberg R, Mims R, Choudhury A, Espinosa C, Zelmanovich J, Okoye NC, Choi EJ, Marder G, Narain S, Gregersen PK, Mackay M, Diamond B, Levy T, Zanos TP, Khosroshahi A, Sanz I, Luning Prak ET, Bar-Or A, Merrill J, Arriens C, Pardo G, Guthridge J, James J, Payne A, Utz PJ, Boss JM, Aranow C, Davidson A. Factors associated with immune responses to SARS-CoV-2 vaccination in individuals with autoimmune diseases. JCI Insight 2024; 9:e180750. [PMID: 38833310 PMCID: PMC11383356 DOI: 10.1172/jci.insight.180750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024] Open
Abstract
Patients with autoimmune diseases are at higher risk for severe infection due to their underlying disease and immunosuppressive treatments. In this real-world observational study of 463 patients with autoimmune diseases, we examined risk factors for poor B and T cell responses to SARS-CoV-2 vaccination. We show a high frequency of inadequate anti-spike IgG responses to vaccination and boosting in the autoimmune population but minimal suppression of T cell responses. Low IgG responses in B cell-depleted patients with multiple sclerosis (MS) were associated with higher CD8 T cell responses. By contrast, patients taking mycophenolate mofetil (MMF) exhibited concordant suppression of B and T cell responses. Treatments with highest risk for low anti-spike IgG response included B cell depletion within the last year, fingolimod, and combination treatment with MMF and belimumab. Our data show that the mRNA-1273 (Moderna) vaccine is the most effective vaccine in the autoimmune population. There was minimal induction of either disease flares or autoantibodies by vaccination and no significant effect of preexisting anti-type I IFN antibodies on either vaccine response or breakthrough infections. The low frequency of breakthrough infections and lack of SARS-CoV-2-related deaths suggest that T cell immunity contributes to protection in autoimmune disease.
Collapse
Affiliation(s)
- Erik Anderson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Michael Powell
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Emily Yang
- Division of Immunology and Rheumatology, Department of Medicine, and
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Kar
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
| | - Tung Ming Leung
- Biostatistics Unit, Office of Academic Affairs, Northwell, New Hyde Park, New York, USA
| | - Cristina Sison
- Biostatistics Unit, Office of Academic Affairs, Northwell, New Hyde Park, New York, USA
| | - Rebecca Steinberg
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
| | - Raven Mims
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ananya Choudhury
- Division of Immunology and Rheumatology, Department of Medicine, and
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, USA
| | - Carlo Espinosa
- Division of Immunology and Rheumatology, Department of Medicine, and
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, USA
| | - Joshua Zelmanovich
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Nkemakonam C. Okoye
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Eun Jung Choi
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Galina Marder
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Sonali Narain
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Peter K. Gregersen
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Meggan Mackay
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Todd Levy
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
| | - Theodoros P. Zanos
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
| | - Arezou Khosroshahi
- Division of Rheumatology, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Amit Bar-Or
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joan Merrill
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Cristina Arriens
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Gabriel Pardo
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Joel Guthridge
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Judith James
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Aimee Payne
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Paul J. Utz
- Division of Immunology and Rheumatology, Department of Medicine, and
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, USA
| | - Jeremy M. Boss
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Cynthia Aranow
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
12
|
D’Anna SE, Vitale AM, D’Amico G, Caruso Bavisotto C, Ambrosino P, Cappello F, Maniscalco M, Marino Gammazza A. Autoimmunity against Nucleus Ambiguous Is Putatively Possible in Both Long-COVID-19 and Vaccinated Subjects: Scientific Evidence and Working Hypothesis. BIOLOGY 2024; 13:359. [PMID: 38927239 PMCID: PMC11200469 DOI: 10.3390/biology13060359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/13/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024]
Abstract
As reported by the World Health Organization (WHO), about 10-20% of people have experienced mid- to long-term effects following SARS-CoV-2 infection, collectively referred to as post-COVID-19 condition or long-COVID, including some neurovegetative symptoms. Numerous findings have suggested that the onset of these neurovegetative symptoms upon viral infection may be caused by the production of autoantibodies through molecular mimicry phenomena. Accordingly, we had previously demonstrated that 22 of the human proteins sharing putatively immunogenic peptides with SARS-CoV-2 proteins are expressed in the dorsal motor nucleus and nucleus ambiguous. Therefore, if molecular mimicry occurs following severe forms of COVID-19, there could be transitory or permanent damage in some vagal structures, resulting in a lower vagal tone and all the related clinical signs. We investigated the presence of autoantibodies against two proteins of vagal nuclei sharing a peptide with SARS-CoV-2 spike glycoprotein using an immunoassay test on blood obtained from patients with cardiorespiratory symptoms in patients affected by ongoing symptomatic COVID-19 (long-COVID), subjects vaccinated without a history of SARS-CoV-2 infection, and subjects not vaccinated without a history of SARS-CoV-2 infection. Interestingly, putative autoantibodies were present in both long-COVID-19 and vaccinated groups, opening interesting questions about pathogenic mechanisms of the disease.
Collapse
Affiliation(s)
| | - Alessandra Maria Vitale
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.E.D.); (P.A.)
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- National Biodiversity Future Center (NBFC), Piazza Marina 61, 90133 Palermo, Italy
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.E.D.); (P.A.)
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| |
Collapse
|
13
|
Rohokale R, Guo J, Guo Z. Monophosphoryl Lipid A-Rhamnose Conjugates as a New Class of Vaccine Adjuvants. J Med Chem 2024; 67:7458-7469. [PMID: 38634150 PMCID: PMC11081837 DOI: 10.1021/acs.jmedchem.3c02385] [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] [Indexed: 04/19/2024]
Abstract
Adjuvant is an integral part of all vaccine formulations but only a few adjuvants with limited efficacies or application scopes are available. Thus, developing more robust and diverse adjuvants is necessary. To this end, a new class of adjuvants having α- and β-rhamnose (Rha) attached to the 1- and 6'-positions of monophosphoryl lipid A (MPLA) was designed, synthesized, and immunologically evaluated in mice. The results indicated a synergistic effect of MPLA and Rha, two immunostimulators that function via interacting with toll-like receptor 4 and recruiting endogenous anti-Rha antibodies, respectively. All the tested MPLA-Rha conjugates exhibited potent adjuvant activities to promote antibody production against both protein and carbohydrate antigens. Overall, MPLA-α-Rha exhibited better activities than MPLA-β-Rha, and 6'-linked conjugates were slightly better than 1-linked ones. Particularly, MPLA-1-α-Rha and MPLA-6'-α-Rha were the most effective adjuvants in promoting IgG antibody responses against protein antigen keyhole limpet hemocyanin and carbohydrate antigen sTn, respectively.
Collapse
Affiliation(s)
- Rajendra Rohokale
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Jiatong Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
14
|
Rio P, Caldarelli M, Chiantore M, Ocarino F, Candelli M, Gasbarrini A, Gambassi G, Cianci R. Immune Cells, Gut Microbiota, and Vaccines: A Gender Perspective. Cells 2024; 13:526. [PMID: 38534370 PMCID: PMC10969451 DOI: 10.3390/cells13060526] [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: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
The development of preventive and therapeutic vaccines has played a crucial role in preventing infections and treating chronic and non-communicable diseases, respectively. For a long time, the influence of sex differences on modifying health and disease has not been addressed in clinical and preclinical studies. The interaction of genetic, epigenetic, and hormonal factors plays a role in the sex-related differences in the epidemiology of diseases, clinical manifestations, and the response to treatment. Moreover, sex is one of the leading factors influencing the gut microbiota composition, which could further explain the different predisposition to diseases in men and women. In the same way, differences between sexes occur also in the immune response to vaccines. This narrative review aims to highlight these differences, focusing on the immune response to vaccines. Comparative data about immune responses, vaccine effectiveness, and side effects are reviewed. Hence, the intricate interplay between sex, immunity, and the gut microbiota will be discussed for its potential role in the response to vaccination. Embracing a sex-oriented perspective in research may improve the efficacy of the immune response and allow the design of tailored vaccine schedules.
Collapse
Affiliation(s)
- Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Monica Chiantore
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Francesca Ocarino
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| |
Collapse
|
15
|
Li L, Li Y, Peng Y, Ma G, Wu J. Investigation on the immune effect of a chitosan-based particle-in-oil-in-water emulsion. Int Immunopharmacol 2024; 128:111468. [PMID: 38171055 DOI: 10.1016/j.intimp.2023.111468] [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: 10/09/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024]
Abstract
Particle-in-oil-in-water (P/O/W) multiple emulsion adjuvants introduce particles into the internal water phase of a water-in-oil-in-water emulsion, combining the advantages of both particle and emulsion adjuvants to enhance humoral and cellular immune responses. In this study, we optimized P/O/W multiple emulsion adjuvants. Chitosan, poly (lactic-co-glycolic acid), and aluminum gel were used to prepare the particles, which were introduced into a water-in-oil-in-water emulsion to obtain three P/O/W multiple emulsion adjuvants. The immune enhancement effects and safety of the three adjuvants were compared, and it was proven that the adjuvant with chitosan nanoparticles in the internal water phase had good cellular and humoral immune effects. Simultaneously, the proportion of the internal water phase increased from 13% to 20%, reducing the antigen concentration required for embedding to one-third of the original concentration and expanding the application range of the composite adjuvant.
Collapse
Affiliation(s)
- Lanxin Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yanan Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, PR China; Division of Molecular Science, Graduate School of Science &Engineering Gunma University, Gunma 376-8515, Japan
| | - Yanan Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China; PLA Key Laboratory of Biopharmaceutical Production and Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Jiangsu National Synergetic Innovation Center for Advanced Materials, Nanjing 210008, PR China.
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China; PLA Key Laboratory of Biopharmaceutical Production and Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Jiangsu National Synergetic Innovation Center for Advanced Materials, Nanjing 210008, PR China.
| |
Collapse
|
16
|
Kanuri SH, Sirrkay PJ. Adjuvants in COVID-19 vaccines: innocent bystanders or culpable abettors for stirring up COVID-heart syndrome. Ther Adv Vaccines Immunother 2024; 12:25151355241228439. [PMID: 38322819 PMCID: PMC10846003 DOI: 10.1177/25151355241228439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
COVID-19 infection is a multi-system clinical disorder that was associated with increased morbidity and mortality. Even though antiviral therapies such as Remdesvir offered modest efficacy in reducing the mortality and morbidity, they were not efficacious in reducing the risk of future infections. So, FDA approved COVID-19 vaccines which are widely administered in the general population worldwide. These COVID-19 vaccines offered a safety net against future infections and re-infections. Most of these vaccines contain inactivated virus or spike protein mRNA that are primarily responsible for inducing innate and adaptive immunity. These vaccines were also formulated to contain supplementary adjuvants that are beneficial in boosting the immune response. During the pandemic, clinicians all over the world witnessed an uprise in the incidence and prevalence of cardiovascular diseases (COVID-Heart Syndrome) in patients with and without cardiovascular risk factors. Clinical researchers were not certain about the underlying reason for the upsurge of cardiovascular disorders with some blaming them on COVID-19 infections while others blaming them on COVID-19 vaccines. Based on the literature review, we hypothesize that adjuvants included in the COVID-19 vaccines are the real culprits for causation of cardiovascular disorders. Operation of various pathological signaling events under the influence of these adjuvants including autoimmunity, bystander effect, direct toxicity, anti-phospholipid syndrome (APS), anaphylaxis, hypersensitivity, genetic susceptibility, epitope spreading, and anti-idiotypic antibodies were partially responsible for stirring up the onset of cardiovascular disorders. With these mechanisms in place, a minor contribution from COVID-19 virus itself cannot be ruled out. With that being said, we strongly advocate for careful selection of vaccine adjuvants included in COVID-19 vaccines so that future adverse cardiac disorders can be averted.
Collapse
Affiliation(s)
- Sri Harsha Kanuri
- Research Fellow, Stark Neurosciences Institute, Indiana University School of Medicine, 320 W 15 ST, Indianapolis, IN 46202, USA
| | | |
Collapse
|
17
|
Gabrielli R, Siani A, Smedile G, Rizzo AR, De Vivo G, Accrocca F, Bartoli S. Isolated popliteal artery lesion due to giant cell vasculitis post COVID-19 mRNA vaccine and COVID-19 asymptomatic infection. Vascular 2024; 32:226-230. [PMID: 36200437 PMCID: PMC9535462 DOI: 10.1177/17085381221126234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Giant cell arteritis (GCA) is a rare granulomatous vasculitis, affecting medium and large vessels, usually in old patients. The incidence of GCA has been higher during current COVID-19 pandemia and COVID-19 is recognized for its immune dysregulation. Lower limbs involvement is uncommon but can be limb threatening, resulting in limb loss. METHOD A 43-year-old man presented with a sudden pain in his right calf and foot associated with pallor and hypothermia, and there was objective evidence of ischemia. Symptoms began few days after he received the first dose of a COVID-19 mRNA vaccine and COVID-19 asymptomatic infection 20 days after vaccination. He had no history of any signs of claudication pre-COVID or limb trauma and was very fit.Enhanced computed tomography and magnetic resonance imaging (MRI)suggest diagnosis of popliteal artery cystic adventitial disease. We resected the affected popliteal artery with interposition using a right great saphenous vein graft, through a posterior approach. On the fourth postoperative day, he was discharged.Histopathological examination revealed patchy intramural inflammatory infiltrates composed of lymphocytes and rare multinucleated giant cells at the internal lamina and adventitia consistent with a diagnosis of GCA. CONCLUSION AND RESULT Our case represents the first reported case of isolated popliteal GCA following vaccination with a COVID-19 mRNA vaccine and COVID-19 infection. We propose that the upregulated immune response to the vaccine acted as a trigger for GCA in this patient with predisposing risk factors and recurrent and repetitive microtrauma in popliteal fossa (the patient is a professional runner). Our case suggests the need for further studies about real world incidence of GCA associated vaccination and COVID-19 infection. Currently, data is limited regarding this relationship. We continue to encourage COVID-19 vaccination, even in elderly patients because the benefits of vaccination far outweigh any theoretical risk of immune dysregulation following administration.
Collapse
Affiliation(s)
- Roberto Gabrielli
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Andrea Siani
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Gianluca Smedile
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Anna Rita Rizzo
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Gennaro De Vivo
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Federico Accrocca
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| | - Stefano Bartoli
- Unit of Vascular, Endovascular and Emergency Vascular Surgery, “S. Eugenio” Hospital, Rome, Italy
| |
Collapse
|
18
|
Chang MS, Kim HR, Kim S, Lee CS, Byeon SH, Kim SS, Lee SW, Kim YJ. Noninfectious Uveitis Risk After COVID-19 Vaccination: A Nationwide Retrospective Cohort Study. Am J Ophthalmol 2024; 258:22-31. [PMID: 37739204 DOI: 10.1016/j.ajo.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/10/2023] [Accepted: 09/16/2023] [Indexed: 09/24/2023]
Abstract
PURPOSE To investigate the incidence and risk of noninfectious uveitis (NIU) following COVID-19 vaccination compared with an unvaccinated, uninfected control group. DESIGN Retrospective population-based cohort study. METHODS We included 5,185,153 individuals who received the first vaccine dose in the exposed group and 2,680,164 individuals in the unexposed, uninfected control group. The study observed for 180 days from their index date. Cumulative incidence and risk of NIU following COVID-19 vaccination, and attributable risk factors were assessed. RESULTS Multivariable analysis showed elevated risk of nonanterior NIU within 60 days (hazard ratio [HR] 1.27 [95% confidence interval {CI} 1.03-1.55] and 61-180 days (HR 1.39 [95% CI 1.20-1.62]). Subgroup analysis highlighted an increased risk in females for early and delayed nonanterior uveitis (HR 1.44 [95% CI 1.08-1.92]; HR 1.78 [95% CI 1.43-2.20], respectively). Regardless of the location and onset timing of uveitis, a history of NIU was identified as the most significant risk factor, with a high hazard ratio ranging from 100 to 200. CONCLUSIONS COVID-19 vaccination may modestly increase the risk of nonanterior uveitis especially in females. Despite adjustments, bias may persist in the exposed group, owing to significant differences between unexposed and exposed groups and low incidence of nonanterior uveitis in the unexposed group. Future research should aim to refine these findings by assessing uveitis risk in prior NIU patients and by enlarging the sample size or cohort matching.
Collapse
Affiliation(s)
- Myung Soo Chang
- From the Department of Ophthalmology (M.S.C., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hae Rang Kim
- Department of Ophthalmology (H.R.K.), CHA Bundang Medical Center, CHA University College of Medicine, Seongnam, Republic of Korea
| | - Sunyeup Kim
- Department of Medical AI (S.K.), Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Christopher Seungkyu Lee
- From the Department of Ophthalmology (M.S.C., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suk Ho Byeon
- From the Department of Ophthalmology (M.S.C., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Soo Kim
- From the Department of Ophthalmology (M.S.C., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Won Lee
- and the Department of Precision Medicine (S.W.L.), Sungkyunkwan University School of Medicine, Suwon, Republic of Korea..
| | - Yong Joon Kim
- From the Department of Ophthalmology (M.S.C., C.S.L., S.H.B., S.S.K., Y.J.K.), Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Primorac Padjen E, Marcec R, Zidar M, Padjen I, Katanec T, Anic B, Likic R. Comparison of reporting rates of arthritis and arthralgia following AstraZeneca, Pfizer-BioNTech, Moderna, and Janssen vaccine administration against SARS-CoV-2 in 2021: analysis of European pharmacovigilance large-scale data. Rheumatol Int 2024; 44:273-281. [PMID: 38142450 DOI: 10.1007/s00296-023-05512-1] [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: 10/15/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023]
Abstract
This study aimed to investigate the reporting rates of arthritis and arthralgia following the administration of four vaccines against SARS-CoV-2: Pfizer-BioNTech (Tozinameran), Moderna (CX-024414), AstraZeneca (Chadox1 NCOV-19), and Janssen (AD26.COV2.S) in 2021. We used data from the EudraVigilance database, specifically analyzing spontaneous reports of suspected adverse reactions (ADRs) from the European Union (EU)/European Economic Area (EEA) region. Age-group-specific reporting rates were calculated by dividing the number of arthralgia and arthritis reports per 1,000,000 vaccine doses administered per age group. Reporting rates were compared using a rate ratio among the four vaccines, using the AstraZeneca vaccine as a comparator. The AstraZeneca vaccine was associated with the highest rate of arthralgia across all age groups. Arthritis reporting rates were significantly lower, with the AstraZeneca vaccine having the highest rates in most age groups, except the 60-69 and 80+ groups, where the Janssen and Pfizer-BioNTech vaccines demonstrated higher reporting rates, respectively. The distribution of arthritis rates did not follow the arthralgia pattern, being higher in the 50-79 age group. This study is the first spontaneous reporting system analysis of arthritis reporting rates post-SARS-CoV-2 vaccination at a European level, revealing a higher reporting of suspected musculoskeletal adverse reactions after AstraZeneca vaccination. The findings underscore the need to consider commonly reported events like arthralgia in risk-benefit assessments prior to vaccination against SARS-CoV-2. Given the high prevalence of rheumatic and musculoskeletal diseases and vaccine hesitancy in this population, our results could influence vaccine choice and acceptance.
Collapse
Affiliation(s)
| | - Robert Marcec
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Matija Zidar
- Faculty of Electrical Engineering and Computing, University of Zagreb, Zagreb, Croatia
| | - Ivan Padjen
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Tomislav Katanec
- School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Branimir Anic
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Robert Likic
- School of Medicine, University of Zagreb, Zagreb, Croatia.
- Division of Clinical Pharmacology and Therapeutics, Department of Internal Medicine, University Hospital Centre Zagreb, Kispaticeva 12, 10000, Zagreb, Croatia.
| |
Collapse
|
20
|
Ning F, Cao XQ, Wang QQ, Li ZY, Ruan Z, Chang T. Safety of SARS-CoV-2 vaccine in patients with autoimmune neurological conditions: A systematic review and meta-analysis. Heliyon 2024; 10:e23944. [PMID: 38261862 PMCID: PMC10796982 DOI: 10.1016/j.heliyon.2023.e23944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/06/2023] [Accepted: 12/16/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Risk of adverse effects and exacerbation in autoimmune neurological conditions (ANC)are frequently cited reasons for COVID-19 vaccine hesitancy. This study evaluates the ANC safety of COVID-19 vaccines in the real world. Methods Electronic databases were searched to identify studies reporting the use of the COVID-19 vaccine in ANC. We selected studies that provided data on adverse effects and worsening conditions related to ANC after vaccination. The pooled incidence rates for various adverse effects, stratified for the disease category, dosage, and type of vaccine, were estimated. Results Twenty-eight studies (31 vaccination cohorts) were included. The pooled incidence rate of general adverse events was 0.35 (95%CI, 0.27-0.43, I2 = 100 %). The pooled incidence rates of local injection reaction, fatigue, weakness, myalgia, fever, headache, and chills were 0.27 (0.18-0.36, I2 = 98 %), 0.16(0.11-0.21, I2 = 93 %), 0.15(0.00-0.31, I2 = 97 %), 0.13(0.08-0.19, I2 = 97 %), 0.11(0.07-0.15, I2 = 95 %), 0.11(0.07-0.16, I2 = 97 %), and 0.09 (0.03-0.16, I2 = 96 %), respectively. The pooled incidence rate of exacerbation adverse events was 0.05 (95%CI, 0.04-0.07, I2 = 84 %). Conclusion According to available evidence, the administration of COVID-19 vaccines in individuals with autoimmune neurological disorders seems well-tolerated, with few reports of adverse events. Furthermore, exacerbation of autoimmune neurological conditions following vaccination appears to be infrequent.
Collapse
Affiliation(s)
| | | | - Qing-qing Wang
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhu-yi Li
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhe Ruan
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | |
Collapse
|
21
|
Negretti GS, Zeiger JS, Cherkas E, Shields CL. Posterior scleritis following COVID-19 vaccination or infection simulating uveal melanoma in 8 consecutive patients. Eye (Lond) 2024; 38:185-191. [PMID: 37422535 PMCID: PMC10764359 DOI: 10.1038/s41433-023-02656-z] [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: 01/10/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/10/2023] Open
Abstract
OBJECTIVES To determine clinical features and outcomes of posterior scleritis masquerading as uveal melanoma following vaccination against COVID-19 and/or COVID-19 infection. SUBJECTS/METHODS All patients with posterior scleritis referred to our service to rule out intraocular tumour between February 2021 and June 2022, who previously had COVID-19 vaccination and/or infection (n = 8). A retrospective detailed review of patient charts and imaging was carried out. RESULTS Previous COVID-19 vaccination was documented in 6 patients (75%) and previous COVID-19 infection and vaccination in 2 patients (25%). Demographic features included mean age of 59 years (median 68, range 5-86 years), white race (n = 7, 87%), and male sex (n = 5, 63%). Mean visual acuity at presentation was 0.24 LogMAR (median 0.18, range 0.0-0.70). The main presenting symptom was blurred vision with pain (n = 5, 63%). Features that suggested scleritis and not uveal melanoma included pain (n = 6, 75%), anterior scleritis (n = 3, 38%), disc oedema (n = 1, 13%), choroidal detachment (n = 3, 38%), choroidal folds (n = 3, 38%), diffusely thickened scleral wall on ultrasonography (n = 2, 25%), Tenon's oedema (n = 5, 63%), and scleral nodule with medium/high internal reflectivity on ultrasonography (n = 4, 50%). Follow-up information at mean of 2 months (range 0.25-7 months) revealed visual acuity at date last seen was mean 0.30 LogMAR (median 0.29, range 0.0-0.54). By 2 months, resolution of "tumour" was noted in 5/6 (83%) patients with follow-up. CONCLUSIONS Posterior scleritis following COVID-19 vaccination and/or infection can masquerade as choroidal melanoma. At 2 months duration, partial or complete resolution of features with minimal visual consequence was noted.
Collapse
Affiliation(s)
- Guy S Negretti
- Shields and Shields, MD, PC, Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jennifer S Zeiger
- Shields and Shields, MD, PC, Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elliot Cherkas
- Shields and Shields, MD, PC, Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
- Casey Eye Institute, OHSU, Portland, OR, USA
| | - Carol L Shields
- Shields and Shields, MD, PC, Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Demidova A, Douguet L, Fert I, Wei Y, Charneau P, Majlessi L. Comparison of preclinical efficacy of immunotherapies against HPV-induced cancers. Expert Rev Vaccines 2024; 23:674-687. [PMID: 38978164 DOI: 10.1080/14760584.2024.2374287] [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: 01/30/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION Persistent infections with the human papilloma viruses, HPV16 and HPV18, are associated with multiple cancers. Although prophylactic vaccines that induce HPV-neutralizing antibodies are effective against primary infections, they have no effect on HPV-mediated malignancies against which there is no approved immuno-therapy. Active research is ongoing in the immunotherapy of these cancers. AREAS COVERED In this review, we compared the preclinical efficacy of vaccine platforms used to treat HPV-induced tumors in the standard model of mice grafted with TC-1 cells, which express the HPV16 E6 and E7 oncoproteins. We searched for the key words, 'HPV,' 'vaccine,' 'therapy,' 'E7,' 'tumor,' 'T cells', and 'mice' for the period from 2005 to 2023 in PubMed and found 330 publications. Among them, we selected the most relevant to extract preclinical antitumor results to enable cross-sectional comparison of their efficacy. EXPERT OPINION SECTION We compared these studies for HPV antigen design, immunization regimen, immunogenicity, and antitumor effect, considering their drawbacks and advantages. Among all strategies used in murine models, certain adjuvanted proteins and viral vectors showed the strongest antitumor effects, with the use of lentiviral vectors being the only approach to result in complete tumor eradication in 100% of experimental individuals while providing the longest-lasting memory.
Collapse
Affiliation(s)
- Anastasia Demidova
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | - Laëtitia Douguet
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | - Ingrid Fert
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | - Yu Wei
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | | |
Collapse
|
23
|
Delkash P, Azimi A, Taherpour N, Haji Aghajani S. The Role of Sinopharm BIBP COVID-19 Vaccine Immunization in Systemic Lupus Erythematous Flare-up. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2023; 18. [DOI: 10.5812/archcid-139989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/22/2023] [Accepted: 12/03/2023] [Indexed: 01/03/2025]
Abstract
Background: This study aimed to investigate the incidence and clinical characteristics of flare-ups in patients with systemic lupus erythematosus (SLE) following immunization with inactivated SARS-CoV-2 vaccines. Methods: In this cross-sectional study at Imam Hossein Hospital's Rheumatology Clinic (Iran), we investigated 72 SLE patients in remission who received the Sinopharm BIBP inactivated COVID-19 vaccine. Their post-vaccination status was monitored for 3 months using the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) checklist by an internal medicine specialist. Results: Fourteen patients (19.44%) experienced symptom flare-ups after vaccination. The most common symptoms were arthritis (64.29%) and skin rash (21.43%). Age, sex, organ involvement, and treatment regimen did not significantly differ between those with and without symptom recurrence (P > 0.05). The second vaccine dose led to more flare-ups compared to the first dose (12.12% vs. 8.33%, P < 0.001). However, the severity of symptom recurrence, measured by the SLEDAI-2K score (P = 0.763), and the interval from vaccination to symptom recurrence (P = 0.075) did not significantly differ between the 2 groups. Except for 2 patients, none of the participants required hospitalization, and flare-up symptoms were effectively managed by prednisolone dosage adjustments. For these 2 patients, the treatment regimen was changed, and the steroid dose was increased; one of them was admitted to the hospital, and the other one was managed on an outpatient basis. Conclusions: The incidence of flare-ups in SLE patients in remission following COVID-19 vaccination with Sinopharm BIBP vaccine was low; most of them were mild and did not require hospitalization, except for 1 patient who was hospitalized after the first dose of vaccination and received rituximab due to vasculitis flare. These findings highlight vaccine safety and underscore the importance of close monitoring, especially after the second dose.
Collapse
|
24
|
Barbey C, Wolf H, Wagner R, Pauly D, Breunig M. A shift of paradigm: From avoiding nanoparticular complement activation in the field of nanomedicines to its exploitation in the context of vaccine development. Eur J Pharm Biopharm 2023; 193:119-128. [PMID: 37838145 DOI: 10.1016/j.ejpb.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
The complement system plays a central role in our innate immunity to fight pathogenic microorganisms, foreign and altered cells, or any modified molecule. Consequences of complement activation include cell lysis, release of histamines, and opsonization of foreign structures in preparation for phagocytosis. Because nanoparticles interact with the immune system in various ways and can massively activate the complement system due to their virus-mimetic size and foreign texture, detrimental side effects have been described after administration like pro-inflammatory responses, inflammation, mild to severe anaphylactic crisis and potentially complement activated-related pseudoallergy (CARPA). Therefore, application of nanotherapeutics has sometimes been observed with restraint, and avoiding or even suppressing complement activation has been of utmost priority. In contrast, in the field of vaccine development, particularly protein-based immunogens that are attached to the surface of nanoparticles, may profit from complement activation regarding breadth and potency of immune response. Improved transport to the regional lymph nodes, enhanced antigen uptake and presentation, as well as beneficial effects on immune cells like B-, T- and follicular dendritic cells may be exploited by strategic nanoparticle design aimed to activate the complement system. However, a shift of paradigm regarding complement activation by nanoparticular vaccines can only be achieved if these beneficial effects are accurately elicited and overshooting effects avoided.
Collapse
Affiliation(s)
- Clara Barbey
- Department of Pharmaceutical Technology, University Regensburg, Regensburg, Germany
| | - Hannah Wolf
- Department of Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Diana Pauly
- Department of Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
25
|
Cheng WJ, Cai ZX, Tang XJ. Adverse reactions to cosmetic implants after COVID-19 vaccination: A literature review. J Cosmet Dermatol 2023; 22:3199-3212. [PMID: 37592436 DOI: 10.1111/jocd.15828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/08/2023] [Accepted: 05/09/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND As the world's population of people vaccinated with the COVID-19 vaccine increases, adverse reactions are increasingly being reported. There have been progressive reports of the effects of COVID-19 vaccination on cosmetic fillers or prostheses, but they have not been reviewed based on their clinical morphologic patterns. This article reviewed the progress of research on adverse reactions to cosmetic implants after COVID-19 vaccination. METHODS We researched the English-language literature up to October 15, 2022, using predefined keywords to identify relevant studies about adverse reactions to cosmetic implants after the COVID-19 vaccination, collecting patient characteristics, implant type, the time interval between vaccination and implantation or injection, time of onset, symptoms, treatments, and outcomes. RESULTS Among the adverse reactions to implants associated with COVID-19 vaccination, we distinguished between (1) injectable fillers and (2) surgical prosthetic implants. The most common adverse reactions were at the site of hyaluronic acid injection and breast prosthesis after Pfizer vaccination, mainly DIRs, and mainly manifested as edema, rash, fever, and capsular contracture. This paper also reported the possible causes, treatments of DIRs, and limitations of current studies. CONCLUSIONS In this article, we attempted to investigate and discuss all the adverse reactions of cosmetic implants related to COVID-19 vaccination in the current literature, to unmask these reactions and make a more accurate assessment of vaccine safety.
Collapse
Affiliation(s)
- Wen-Jie Cheng
- Department of Craniomaxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zi-Xing Cai
- Xiamen University Medical College, Fujian, China
| | - Xiao-Jun Tang
- Department of Craniomaxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Sacchi MC, Pelazza C, Bertolotti M, Agatea L, De Gaspari P, Tamiazzo S, Ielo D, Stobbione P, Grappiolo M, Bolgeo T, Novel P, Ciriello MM, Maconi A. The onset of de novo autoantibodies in healthcare workers after mRNA based anti-SARS-CoV-2 vaccines: a single centre prospective follow-up study. Autoimmunity 2023; 56:2229072. [PMID: 37381619 DOI: 10.1080/08916934.2023.2229072] [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: 12/21/2022] [Revised: 06/14/2023] [Accepted: 06/18/2023] [Indexed: 06/30/2023]
Abstract
Nowadays, data concerning the risk of autoimmune disease after SARS-CoV-2 (COVID-19) vaccination is controversial. The aim of this single centre prospective follow-up study was to evaluate whether healthcare workers (HCWs) vaccinated with BNT162b2 mRNA and mRNA-1273 will show a development and/or a persistence of autoantibodies, focussing on the detection of antibodies against nuclear antigens (antinuclear antibodies, ANA). We enrolled 155 HCWs, however only 108 of them received the third dose and were considered for further analysis. Blood samples were collected before vaccine inoculation (T0), at 3 (T1) and 12 months (T2) after the first dose. All samples were analysed for the presence of a) ANA using indirect Immunofluorescence [IIF] (dilutions of 1:80, 1:160. 1:320 and 1:640), and anti-smooth muscle antibodies (ASMA); b) anti-myeloperoxidase (anti-MPO), anti-proteinase 3 (anti-PR3) and anti-citrullinated peptide antibodies (aCCP) [FEIA]; c) anti-phospholipid antibodies (anticardiolipin [aCL], anti-beta-2- glycoprotein I [anti-ß-2GPI] (Chemiluminescence). Line-blot technology was performed using the following kit: EUROLINE ANA profile 3 plus DFS70 (IgG). Our research suggests that mRNA based anti-SARSCoV-2 vaccines can induce the production of de novo ANA in 22/77(28,57%) of subjects and that the percentage of positivity seems to be directly correlated to the number of vaccine expositions: 6/77 (7,79%) after 2 doses; 16/77 (20,78%) after 3 doses. Since it is known that hyperstimulation of the immune system could lead to autoimmunity, these preliminary results seem to further sustain the idea that the hyperstimulation of the immune system might lead to an autoinflammatory mechanism and eventually to autoimmune disorders. However, the link between SARS-CoV-2 vaccination and the development of autoimmune diseases needs to be further investigated.
Collapse
Affiliation(s)
- M C Sacchi
- Autoimmunology and Analysis Laboratory Unit, "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
- Research Laboratory Facility, Research and Innovation Department (DAIRI), "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - C Pelazza
- Research Training Innovation Infrastructure, Research and Innovation Department (DAIRI), "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - M Bertolotti
- Research Training Innovation Infrastructure, Research and Innovation Department (DAIRI), "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - L Agatea
- Laboratory Department, Affiliated to Euroimmun, Padova, Italy
| | - P De Gaspari
- Laboratory Department, Affiliated to Euroimmun, Padova, Italy
| | - S Tamiazzo
- Autoimmunology and Analysis Laboratory Unit, "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - D Ielo
- Werfen, EEMEA, Milan, Italy
| | - P Stobbione
- Rheumatology Unit, "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - M Grappiolo
- Autoimmunology and Analysis Laboratory Unit, "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - T Bolgeo
- Research Training Innovation Infrastructure, Research and Innovation Department (DAIRI), "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - P Novel
- Laboratory Department, Affiliated to Euroimmun, Padova, Italy
| | - M M Ciriello
- Autoimmunology and Analysis Laboratory Unit, "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - A Maconi
- Research and Innovation Department (DAIRI), "SS. Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| |
Collapse
|
27
|
Kupani M, Pandey RK, Vashisht S, Singh S, Prajapati VK, Mehrotra S. Prediction of an immunogenic peptide ensemble and multi-subunit vaccine for Visceral leishmaniasis using bioinformatics approaches. Heliyon 2023; 9:e22121. [PMID: 38196838 PMCID: PMC10775901 DOI: 10.1016/j.heliyon.2023.e22121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 01/11/2024] Open
Abstract
Visceral Leishmaniasis (VL) is a neglected tropical disease of public health importance in the Indian subcontinent. Despite consistent elimination initiatives, the disease has not yet been eliminated and there is an increased risk of resurgence from active VL reservoirs including asymptomatic, post kala azar dermatitis leishmaniasis (PKDL) and HIV-VL co-infected individuals. To achieve complete elimination and sustain it in the long term, a prophylactic vaccine, which can elicit long lasting immunity, is desirable. In this study, we employed immunoinformatic tools to design a multi-subunit epitope vaccine for the Indian population by targeting antigenic secretory proteins screened from the Leishmania donovani proteome. Out of 8014 proteins, 277 secretory proteins were screened for their cellular location and proteomic evidence. Through NCBI BlastP, unique fragments of the proteins were cropped, and their antigenicity was evaluated. B-cell, HTL and CTL epitopes as well as IFN-ɣ, IL-17, and IL-10 inducers were predicted, manually mapped to the fragments and common regions were tabulated forming a peptide ensemble. The ensemble was evaluated for Class I MHC immunogenicity and toxicity. Further, immunogenic peptides were randomly selected and used to design vaccine constructs. Eight vaccine constructs were generated by linking random peptides with GS linkers. Synthetic TLR-4 agonist, RS09 was used as an adjuvant and linked with the constructs using EAAK linkers. The predicted population coverage of the constructs was ∼99.8 % in the Indian as well as South Asian populations. The most antigenic, nontoxic, non-allergic construct was chosen for the prediction of secondary and tertiary structures. The 3D structures were refined and analyzed using Ramachandran plot and Z-scores. The construct was docked with TLR-4 receptor. Molecular dynamic simulation was performed to check for the stability of the docked complex. Comparative in silico immune simulation studies showed that the predicted construct elicited humoral and cell-mediated immunity in human host comparable to that elicited by Leish-F3, which is a promising vaccine candidate for human VL.
Collapse
Affiliation(s)
- Manu Kupani
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Rajeev Kumar Pandey
- Research & Development, Thermo Fisher Scientific, Bangalore, 560066, Karnataka, India
| | - Sharad Vashisht
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, 121001, Harayana, India
| | - Satyendra Singh
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| |
Collapse
|
28
|
Aikawa NE, Borba EF, Balbi VA, Sallum AME, Buscatti IM, Campos LMA, Kozu KT, Garcia CC, Capão ASV, de Proença ACT, Leon EP, da Silva Duarte AJ, Lopes MH, Silva CA, Bonfá E. Safety and immunogenicity of influenza A(H3N2) component vaccine in juvenile systemic lupus erythematosus. Adv Rheumatol 2023; 63:55. [PMID: 38017564 DOI: 10.1186/s42358-023-00339-7] [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: 08/15/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023] Open
Abstract
INTRODUCTION Seasonal influenza A (H3N2) virus is an important cause of morbidity and mortality in the last 50 years in population that is greater than the impact of H1N1. Data assessing immunogenicity and safety of this virus component in juvenile systemic lupus erythematosus (JSLE) is lacking in the literature. OBJECTIVE To evaluate short-term immunogenicity and safety of influenza A/Singapore (H3N2) vaccine in JSLE. METHODS 24 consecutive JSLE patients and 29 healthy controls (HC) were vaccinated with influenza A/Singapore/INFIMH-16-0019/2016(H3N2)-like virus. Influenza A (H3N2) seroprotection (SP), seroconversion (SC), geometric mean titers (GMT), factor increase in GMT (FI-GMT) titers were assessed before and 4 weeks post-vaccination. Disease activity, therapies and adverse events (AE) were also evaluated. RESULTS JSLE patients and controls were comparable in current age [14.5 (10.1-18.3) vs. 14 (9-18.4) years, p = 0.448] and female sex [21 (87.5%) vs. 19 (65.5%), p = 0.108]. Before vaccination, JSLE and HC had comparable SP rates [22 (91.7%) vs. 25 (86.2%), p = 0.678] and GMT titers [102.3 (95% CI 75.0-139.4) vs. 109.6 (95% CI 68.2-176.2), p = 0.231]. At D30, JSLE and HC had similar immune response, since no differences were observed in SP [24 (100%) vs. 28 (96.6%), p = 1.000)], SC [4 (16.7%) vs. 9 (31.0%), p = 0.338), GMT [162.3 (132.9-198.3) vs. 208.1 (150.5-287.8), p = 0.143] and factor increase in GMT [1.6 (1.2-2.1) vs. 1.9 (1.4-2.5), p = 0.574]. SLEDAI-2K scores [2 (0-17) vs. 2 (0-17), p = 0.765] and therapies remained stable throughout the study. Further analysis of possible factors influencing vaccine immune response among JSLE patients demonstrated similar GMT between patients with SLEDAI < 4 compared to SLEDAI ≥ 4 (p = 0.713), as well as between patients with and without current use of prednisone (p = 0.420), azathioprine (p = 1.0), mycophenolate mofetil (p = 0.185), and methotrexate (p = 0.095). No serious AE were reported in both groups and most of them were asymptomatic (58.3% vs. 44.8%, p = 0.958). Local and systemic AE were alike in both groups (p > 0.05). CONCLUSION This is the first study that identified adequate immune protection against H3N2-influenza strain with additional vaccine-induced increment of immune response and an adequate safety profile in JSLE. ( www. CLINICALTRIALS gov , NCT03540823).
Collapse
Affiliation(s)
- Nadia Emi Aikawa
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil.
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - Eduardo Ferreira Borba
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Verena Andrade Balbi
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
| | - Adriana Maluf Elias Sallum
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
| | - Izabel Mantovani Buscatti
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
| | - Lucia Maria Arruda Campos
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
| | - Kátia Tomie Kozu
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
| | - Cristiana Couto Garcia
- Laboratory of Respiratory, Exanthematic Viruses, Enterovirus and Viral Emergencies, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
- Integrated Research Group On Biomarkers. René Rachou Institute, FIOCRUZ Minas, Belo Horizonte, MG, Brazil
| | - Artur Silva Vidal Capão
- Laboratory of Respiratory, Exanthematic Viruses, Enterovirus and Viral Emergencies, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Adriana Coracini Tonacio de Proença
- Department of Infectious and Parasitic Diseases, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Elaine Pires Leon
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alberto José da Silva Duarte
- Clinical Laboratory Division - Department of Pathology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Marta Heloisa Lopes
- Department of Infectious and Parasitic Diseases, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Clovis Artur Silva
- Pediatric Rheumatology Unit, Instituto da Criança e do Adolescente, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, 3Rd Floor, room 3190 - Cerqueira Cesar, São Paulo, SP, CEP 05403-010, Brazil
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Eloisa Bonfá
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
29
|
Russi RC, del Balzo D, Reidel IG, Alonso Bivou M, Flor N, Lujan A, Sanchez D, Damiani MT, Veaute C. Evaluation of three formulations based on Polymorphic membrane protein D in mice infected with Chlamydia trachomatis. Front Immunol 2023; 14:1267684. [PMID: 38045697 PMCID: PMC10690417 DOI: 10.3389/fimmu.2023.1267684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
The significant impact of Chlamydia trachomatis(Ct) infections worldwide highlights the need to develop a prophylactic vaccine that elicits effective immunity and protects the host from the immunopathological effects of Ct infection. The aim of this study was to evaluate a vaccine based on a fragment of the Polymorphic membrane protein D (FPmpD) of C. trachomatis as an immunogen using a heterologous DNA prime-protein boost strategy in female mice Three different formulations were evaluated as protein boost: free recombinant FPmpD (rFPmpD) or rFPmpD formulated with a liposomal adjuvant alternatively supplemented with CpG or a cationic gemini lipopeptide as immunostimulants. The three candidates induced an increase in the cervicovaginal and systemic titers of anti-rFPmpD antibodies in two strains of mice (BALB/c and C57BL/6), with no evidence of fertility alterations. The three formulations induced a rapid and robust humoral immune response upon the Ct challenge. However, the booster with free rFPmpD more efficiently reduced the shedding of infective Ct and prevented the development of immunopathology. The formulations containing adjuvant induced a strong inflammatory reaction in the uterine tissue. Hence, the prime-boost strategy with the adjuvant-free FPmpD vaccine formulation might constitute a promissory candidate to prevent C. trachomatis intravaginal infection.
Collapse
Affiliation(s)
- Romina Cecilia Russi
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Diego del Balzo
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ivana Gabriela Reidel
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Mariano Alonso Bivou
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Noelia Flor
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Agustín Lujan
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Sanchez
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Teresa Damiani
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Carolina Veaute
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| |
Collapse
|
30
|
Zhu J, Liu J, Yan C, Wang D, Pan W. Trained immunity: a cutting edge approach for designing novel vaccines against parasitic diseases? Front Immunol 2023; 14:1252554. [PMID: 37868995 PMCID: PMC10587610 DOI: 10.3389/fimmu.2023.1252554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The preventive situation of parasitosis, a global public health burden especially for developing countries, is not looking that good. Similar to other infections, vaccines would be the best choice for preventing and controlling parasitic infection. However, ideal antigenic molecules for vaccine development have not been identified so far, resulting from the complicated life history and enormous genomes of the parasites. Furthermore, the suppression or down-regulation of anti-infectious immunity mediated by the parasites or their derived molecules can compromise the effect of parasitic vaccines. Comparing the early immune profiles of several parasites in the permissive and non-permissive hosts, a robust innate immune response is proposed to be a critical event to eliminate the parasites. Therefore, enhancing innate immunity may be essential for designing novel and effective parasitic vaccines. The newly emerging trained immunity (also termed innate immune memory) has been increasingly recognized to provide a novel perspective for vaccine development targeting innate immunity. This article reviews the current status of parasitic vaccines and anti-infectious immunity, as well as the conception, characteristics, and mechanisms of trained immunity and its research progress in Parasitology, highlighting the possible consideration of trained immunity in designing novel vaccines against parasitic diseases.
Collapse
Affiliation(s)
- Jinhang Zhu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, Zhejiang, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
31
|
Lee Y, Jeong M, Park J, Jung H, Lee H. Immunogenicity of lipid nanoparticles and its impact on the efficacy of mRNA vaccines and therapeutics. Exp Mol Med 2023; 55:2085-2096. [PMID: 37779140 PMCID: PMC10618257 DOI: 10.1038/s12276-023-01086-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 10/03/2023] Open
Abstract
Several studies have utilized a lipid nanoparticle delivery system to enhance the effectiveness of mRNA therapeutics and vaccines. However, these nanoparticles are recognized as foreign materials by the body and stimulate innate immunity, which in turn impacts adaptive immunity. Therefore, it is crucial to understand the specific type of innate immune response triggered by lipid nanoparticles. This article provides an overview of the immunological response in the body, explores how lipid nanoparticles activate the innate immune system, and examines the adverse effects and immunogenicity-related development pathways associated with these nanoparticles. Finally, we highlight and explore strategies for regulating the immunogenicity of lipid nanoparticles.
Collapse
Affiliation(s)
- Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Jeongeun Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyein Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea.
| |
Collapse
|
32
|
Fiorelli D, Caruso V, Belardi R, Bernardini S, Nuccetelli M. Evaluation of autoantibody profile in healthy subjects after mRNA vaccination against COVID-19. Int Immunopharmacol 2023; 122:110592. [PMID: 37413933 DOI: 10.1016/j.intimp.2023.110592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/16/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND SARS-CoV-2 severe acute respiratory syndrome has rapidly spread worldwide since 2019. All scientific and technological forces have concentrated towards the formulation of vaccines to contain the disease. In less than one year (December 2020) a first messenger RNA vaccine (Comirnaty, BioNTech/Pfizer) was authorized. However, the research community has wondered about possible side effects on the immune system, given the vaccines administration in phase 4. AIM This study aims to evaluate the mRNA vaccine impact on the development of possible positive autoantibody profile in healthcare workers without any previous underlying pathology, after first, second and booster dose of Pfizer vaccine, by determining: circulating immune complexes concentrations (CIC); anti-myeloperoxidase (MPO) and anti-proteinase 3 (PR3) autoantibodies, the presence of antinuclear antibodies (ANA) and subsequent second level tests (extractable nuclear antigen (ENA) screen, double-strand DNA, extractable nuclear antigen (ANA) profile). METHODS The subjects were divided according to anti-SARS-CoV-2 IgG RBD antibodies increasing concentrations in: Group I < 10 BAU/ml (N = 114); Group II > 1000 BAU/ml (N = 112); Group III > 2500 BAU/ml (N = 78). RESULTS Our data show no autoreactive response changes over time in healthy subjects after vaccination. In fact, evaluation of ANA, CIC, anti-MPO, anti-PR3 and the detection of specific autoantigens, did not display significant variations. CONCLUSIONS The results suggest the exclusion of a correlation between the administration of the vaccine and the possible onset of autoimmune disorders. Nevertheless, further investigations will be needed to test for any long-term side effects on an ever-growing population.
Collapse
Affiliation(s)
- Denise Fiorelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy.
| | - Vincenza Caruso
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Riccardo Belardi
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| |
Collapse
|
33
|
Pasoto SG, Borba EF, Formiga FFC, do Nascimento Pedrosa T, Aikawa NE, de Siqueira MAMT, Capão ASV, de Proença ACT, Fuller R, Yuki EFN, Leon EP, de Oliveira Martins VA, Lopes MH, da Silva Duarte AJ, da Silva CAA, Bonfa E. Robust immunogenicity to the H3N2 component of influenza A vaccine in primary Sjögren syndrome. Clin Rheumatol 2023; 42:2419-2425. [PMID: 37306813 DOI: 10.1007/s10067-023-06666-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Influenza A (H3N2) virus is the major cause of morbidity/mortality due to seasonal influenza over 50 years. Data about the safety/immunogenicity of influenza A/Singapore (H3N2) vaccine are scarce in primary Sjögren syndrome (pSS). METHODS Twenty-one consecutive pSS patients and 42 HC (healthy control individuals) were immunized with influenza A/Singapore/INFIMH-16-0019/2016 (H3N2)-like virus. Rates of SP (seroprotection) and SC (seroconversion), GMT (geometric mean titers), FI-GMT (factor increase in GMT), ESSDAI (EULAR Sjögren's Syndrome Disease Activity Index), and adverse events were appraised before and 4 weeks post-vaccination. RESULTS pSS and HC had similar mean age (51.2 ± 14.2 vs. 50.6 ± 12.1 years, p = 0.886). Pre-vaccination SP rates were high in pSS and HC (90.5% vs. 71.4%, p = 0.114), and GMT were higher in pSS [80.0 (52.4-160.0) vs. 40.0 (20.0-80.0), p = 0.001]. The percentage of influenza vaccination in the preceding two years was elevated and similar in pSS and HC (94.1% vs. 94.6%, p = 1.000). GMT values augmented in both groups four weeks after vaccination and persisted higher in the first group [160.0 (80.0-320.0) vs. 80.0 (40.0-80.0), p < 0.001] with equivalent FI-GMT [1.4 (1.0-2.8) vs. 1.4 (1.0-2.0), p = 0.410]. Both groups had low and similar SC rates (19.0% vs. 9.5%, p = 0.423). ESSDAI values persisted steadily during the study (p = 0.313). No serious adverse events have occurred. CONCLUSION The novel demonstration that the influenza A/Singapore (H3N2) vaccine induces a different pattern of immunogenicity from other influenza A constituents in pSS, featured by a desirable high pre- and post-vaccination immunogenicity, is in line with reported differences in immune responses between strains in trivalent vaccines and may be related to pre-existing immunity. CLINICALTRIALS gov: #NCT03540823. Key Points • This prospective study demonstrated a robust pre- and post-vaccination immunogenicity to influenza A/Singapore/INFIMH-16-0019/2016 (H3N2)-like virus in primary Sjögren's syndrome (pSS). • This high immunogenicity pattern may be related to pre-existing immunization, or else it is related to immunogenicity differences of each strain. • This vaccine had an adequate safety profile in pSS, with no impact on disease activity.
Collapse
Affiliation(s)
- Sandra Gofinet Pasoto
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
| | - Eduardo Ferreira Borba
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Francisco Fellipe Claudino Formiga
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Tatiana do Nascimento Pedrosa
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Nadia Emi Aikawa
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
- Pediatric Rheumatology Unit, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | | | - Artur Silva Vidal Capão
- Laboratory of Respiratory Virus and Measles, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Adriana Coracini Tonacio de Proença
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Ricardo Fuller
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Emily Figueiredo Neves Yuki
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Elaine Pires Leon
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Victor Adriano de Oliveira Martins
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Marta Heloisa Lopes
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Alberto José da Silva Duarte
- Clinical Laboratory Division, Department of Pathology, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Clovis Artur Almeida da Silva
- Pediatric Rheumatology Unit, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Eloisa Bonfa
- Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| |
Collapse
|
34
|
Xu S, Yu H, Cheng X, Wu J, Bao J, Zhang L. Inactivated SARS-CoV-2 vaccination does not disturb the clinical course of Graves' disease: An observational cohort study. Vaccine 2023; 41:5648-5654. [PMID: 37544826 DOI: 10.1016/j.vaccine.2023.07.053] [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: 01/08/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
SARS-CoV-2 vaccination has been reported to be associated with the induction of thyroid disorders. To investigate the influence of SARS-CoV-2 vaccination on the disease course of patients who were undergoing treatment for Graves' disease (GD), a total of 651 consecutive GD patients who attended follow-up visits in Jiangyuan Hospital were enrolled in this retrospective study, including 443 inactivated SARS-CoV-2 vaccine recipients and 208 unvaccinated participants. The changes in serum levels of free triiodothyronine (fT3), free thyroxine (fT4), thyroid stimulating hormone (TSH) and TSH receptor antibody (TRAb) were analyzed. Crude and adjusted hazard ratios (HRs) were estimated using Cox regression models to investigate the risks in incident TRAb positivity and hyperthyroidism recurrence following vaccination. The median levels of TRAb and fT3 significantly decreased in both vaccinated and unvaccinated groups during the GD hyperthyroidism treatment. The fT4 levels of both groups were well within normal limits and presented downward trends simultaneously. Although the present study observed an increasing trend of TSH level during follow-up, significant difference was not seen in both vaccinated and unvaccinated groups. Except for newly diagnosed GD patients, vaccinated participants had significantly lower risks of incident TRAb positivity (adjusted HR = 0.22; 95%CI: 0.10-0.48, P < 0.001) after adjusted for sex, age, disease duration and MMI dose at baseline. Besides, vaccination was unlikely to serve as a risk factor for hyperthyroidism recurrence (adjusted HR = 1.20; 95%CI: 0.51-2.83, P = 0.678). Notably, newly diagnosed patients who received vaccination were just as likely to achieve remission of thyrotoxicosis as those not receiving the vaccination at any time. Our results concluded that inactivated SARS-CoV-2 vaccination would not disturb the treatment course among GD hyperthyroidism patients.
Collapse
Affiliation(s)
- Shichen Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Xian Cheng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jing Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jiandong Bao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Li Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; School of Life Science and Technology, Southeast University, Nanjing 210096, China.
| |
Collapse
|
35
|
Galiciolli MEA, Silva JF, Prodocimo MM, Laureano HA, Calado SLDM, Oliveira CS, Guiloski IC. Toxicological Effects of Thimerosal and Aluminum in the Liver, Kidney, and Brain of Zebrafish ( Danio rerio). Metabolites 2023; 13:975. [PMID: 37755255 PMCID: PMC10537066 DOI: 10.3390/metabo13090975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/28/2023] Open
Abstract
Vaccination programs in the first years of a child's life are effective and extremely important strategies for the successful eradication of diseases. However, as no intervention is without risks, the metal-based components of some vaccines, such as thimerosal (TMS), a preservative composed of ethylmercury, and aluminum (Al), have begun to generate distrust on the part of the population. Therefore, this study evaluated the effects of exposure to thimerosal and aluminum hydroxide (alone or in mixture) on Danio rerio (zebrafish) specimens. The fish were exposed to thimerosal and/or aluminum hydroxide intraperitoneally. The liver, kidney, and brain were removed for a biochemical biomarker analysis, histopathological analysis, and metal quantification. As a result, we observed changes in the activity of the analyzed enzymes (SOD, GST, GPx) in the kidney and brain of the zebrafish, a reduction in GSH levels in all analyzed tissues, and a reduction in MT levels in the kidney and liver as well as in the brain. Changes in AChE enzyme activity were observed. The biochemical results corroborate the changes observed in the lesion index and histomorphology sections. We emphasize the importance of joint research on these compounds to increase the population's safety against their possible toxic effects.
Collapse
Affiliation(s)
- Maria Eduarda Andrade Galiciolli
- Instituto de Pesquisa Pelé Pequeno Príncipe, Avenida Silva Jardim, 1632, Água Verde, Curitiba 80250-200, PR, Brazil; (M.E.A.G.); (J.F.S.)
- Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Juliana Ferreira Silva
- Instituto de Pesquisa Pelé Pequeno Príncipe, Avenida Silva Jardim, 1632, Água Verde, Curitiba 80250-200, PR, Brazil; (M.E.A.G.); (J.F.S.)
- Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Maritana Mela Prodocimo
- Departamento de Biologia Celular e Molecular, Universidade Federal do Paraná, Centro Politécnico, Avenida Cel. Francisco H. dos Santos, 100—Jardim das Américas, Curitiba—PR, Curitiba 81531-980, PR, Brazil;
| | - Henrique Aparecido Laureano
- Instituto de Pesquisa Pelé Pequeno Príncipe, Avenida Silva Jardim, 1632, Água Verde, Curitiba 80250-200, PR, Brazil; (M.E.A.G.); (J.F.S.)
- Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | | | - Claudia Sirlene Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Avenida Silva Jardim, 1632, Água Verde, Curitiba 80250-200, PR, Brazil; (M.E.A.G.); (J.F.S.)
- Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Izonete Cristina Guiloski
- Instituto de Pesquisa Pelé Pequeno Príncipe, Avenida Silva Jardim, 1632, Água Verde, Curitiba 80250-200, PR, Brazil; (M.E.A.G.); (J.F.S.)
- Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| |
Collapse
|
36
|
Silva AJD, de Sousa MMG, de Macêdo LS, de França Neto PL, de Moura IA, Espinoza BCF, Invenção MDCV, de Pinho SS, da Gama MATM, de Freitas AC. RNA Vaccines: Yeast as a Novel Antigen Vehicle. Vaccines (Basel) 2023; 11:1334. [PMID: 37631902 PMCID: PMC10459952 DOI: 10.3390/vaccines11081334] [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: 07/03/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/28/2023] Open
Abstract
In the last decades, technological advances for RNA manipulation enabled and expanded its application in vaccine development. This approach comprises synthetic single-stranded mRNA molecules that direct the translation of the antigen responsible for activating the desired immune response. The success of RNA vaccines depends on the delivery vehicle. Among the systems, yeasts emerge as a new approach, already employed to deliver protein antigens, with efficacy demonstrated through preclinical and clinical trials. β-glucans and mannans in their walls are responsible for the adjuvant property of this system. Yeast β-glucan capsules, microparticles, and nanoparticles can modulate immune responses and have a high capacity to carry nucleic acids, with bioavailability upon oral immunization and targeting to receptors present in antigen-presenting cells (APCs). In addition, yeasts are suitable vehicles for the protection and specific delivery of therapeutic vaccines based on RNAi. Compared to protein antigens, the use of yeast for DNA or RNA vaccine delivery is less established and has fewer studies, most of them in the preclinical phase. Here, we present an overview of the attributes of yeast or its derivatives for the delivery of RNA-based vaccines, discussing the current challenges and prospects of this promising strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (A.J.D.S.)
| |
Collapse
|
37
|
Bai Y, Niu Y, Qin S, Ma G. A New Biomaterial Derived from Aloe vera-Acemannan from Basic Studies to Clinical Application. Pharmaceutics 2023; 15:1913. [PMID: 37514099 PMCID: PMC10385217 DOI: 10.3390/pharmaceutics15071913] [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: 06/07/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Aloe vera is a kind of herb rich in polysaccharides. Acemannan (AC) is considered to be a natural polysaccharide with good biodegradability and biocompatibility extracted from Aloe vera and has a wide range of applications in the biomedical field due to excellent immunomodulatory, antiviral, antitumor, and tissue regeneration effects. In recent years, clinical case reports on the application of AC as a novel biomedical material in tissue regenerative medicine have emerged; it is mainly used in bone tissue engineering, pulp-dentin complex regeneration engineering, and soft tissue repair, among other operations. In addition, multiple studies have proved that the new composite products formed by the combination of AC and other compounds have excellent biological and physical properties and have broader research prospects. This paper introduces the preparation process, surface structure, and application forms of AC; summarizes the influence of acetyl functional group content in AC on its functions; and provides a detailed review of the functional properties, laboratory studies, clinical cutting-edge applications, and combined applications of AC. Finally, the current application status of AC from basic research to clinical treatment is analyzed and its prospects are discussed.
Collapse
Affiliation(s)
- Yingjie Bai
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, China
| | - Yimeng Niu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, China
| | - Shengao Qin
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, China
| | - Guowu Ma
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, China
- Department of Stomatology, Stomatological Hospital Affiliated School, Stomatology of Dalian Medical University, NO. 397 Huangpu Road, Shahekou District, Dalian 116086, China
| |
Collapse
|
38
|
Seida I, Alrais M, Seida R, Alwani A, Kiyak Z, Elsalti A, Nil Esirgun S, Abali T, Mahroum N. Autoimmune/inflammatory syndrome induced by adjuvants (ASIA): past, present, and future implications. Clin Exp Immunol 2023; 213:87-101. [PMID: 36881788 PMCID: PMC10324553 DOI: 10.1093/cei/uxad033] [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: 10/08/2022] [Revised: 02/06/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023] Open
Abstract
Adjuvants, as the name indicates, are adjoined material aimed to assist in functioning as when added to vaccines they are meant to boost the effect and strongly stimulate the immune system. The response of the immune system can be unpredictable, and the autoimmune/inflammatory syndrome induced by adjuvants (ASIA) was developed to address possible adverse reactions of an autoimmune and inflammatory type that may be caused by adjuvants. While ASIA, as a syndrome, was coined and defined in 2011; reports describing patients with vague and nonspecific clinical symptoms following vaccinations appeared much earlier. In other words, ASIA came to define, arrange, and unite the variety of symptoms, related to autoimmunity, caused not by the vaccine itself, rather by the adjuvant part of the vaccine such as aluminum, among others. Accordingly, the introduction of ASIA enabled better understanding, proper diagnosis, and early treatment of the disorder. Furthermore, ASIA was shown to be associated with almost all body systems and various rheumatic and autoimmune diseases such as systemic lupus erythematosus, antiphospholipid syndrome, and systemic sclerosis. In addition, the correlation between COVID-19 and ASIA was noticed during the pandemic. In this review, we summarized the reported effects of adjuvants and medical literature before and after ASIA was defined, the several ways ASIA can manifest and impact different systems of the body, and the incidences of ASIA during the COVID-19 pandemic. It is important to clarify, that vaccines are among, if not the, most effective means of fighting infectious diseases however, we believe that vaccines manufacturing is not above criticism, particularly when it comes to added substances possessing a risk of side effects.
Collapse
Affiliation(s)
- Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mahmoud Alrais
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulkarim Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zeynep Kiyak
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Sevval Nil Esirgun
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tunahan Abali
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
39
|
Aroffu M, Manca ML, Pedraz JL, Manconi M. Liposome-based vaccines for minimally or noninvasive administration: an update on current advancements. Expert Opin Drug Deliv 2023; 20:1573-1593. [PMID: 38015659 DOI: 10.1080/17425247.2023.2288856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION Vaccination requires innovation to provide effective protection. Traditional vaccines have several drawbacks, which can be overcome with advanced technologies and different administration routes. Over the past 10 years, a significant amount of research has focussed on the delivery of antigens into liposomes due to their dual role as antigen-carrying systems and vaccine adjuvants able to increase the immunogenicity of the carried antigen. AREAS COVERED This review encompasses the progress made over the last 10 years with liposome-based vaccines designed for minimally or noninvasive administration, filling the gaps in previous reviews and providing insights on composition, administration routes, results achieved, and Technology Readiness Level of the most recent formulations. EXPERT OPINION Liposome-based vaccines administered through minimally or noninvasive routes are expected to improve efficacy and complacency of vaccination programs. However, the translation from lab-scale production to large-scale production and collaborations with hospitals, research centers, and companies are needed to allow new products to enter the market and improve the vaccination programs in the future.
Collapse
Affiliation(s)
- Matteo Aroffu
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Maria Letizia Manca
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- BioAraba, NanoBioCel research Group, Vitoria-Gasteiz, Spain
| | - Maria Manconi
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
| |
Collapse
|
40
|
Isoda A, Sairenji Y, Mihara M, Iriuchishima H, Saito A. Coexistence of IgG4-Related Disease and Reactive Granuloma to Paraffin Plombage. Cureus 2023; 15:e40620. [PMID: 37476122 PMCID: PMC10354829 DOI: 10.7759/cureus.40620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/22/2023] Open
Abstract
We present a patient with IgG4-related disease (IgG4-RD) that developed after receiving extra-periosteal paraffin-embedded therapy for the treatment of pulmonary tuberculosis. The patient showed clinicopathological features consistent with IgG4-RD, including the enlargement of affected organs (salivary glands, lymph nodes, and retroperitoneal soft tissue mass), elevation of serum IgG4 levels, and infiltration of IgG4-positive plasma cells. The presence of reactive granulomas with foreign body giant cells (FBGCs) surrounding the paraffin-filled site suggested a type 2 helper T (Th2)-dominant immune response induced by the implanted biomaterial. Furthermore, paraffin, known to act as an adjuvant, may have played a role in activating the immune response and inducing IgG4-RD-like symptoms. This case highlights the potential relationship between foreign substances and the development of autoimmune diseases such as IgG4-RD.
Collapse
Affiliation(s)
- Atsushi Isoda
- Department of Hematology, Iryohojin Hoshiiin, Maebashi, JPN
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, JPN
| | - Yukiko Sairenji
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, JPN
| | - Masahiro Mihara
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, JPN
| | - Hirono Iriuchishima
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, JPN
| | - Akio Saito
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, JPN
| |
Collapse
|
41
|
Tsai TF, Ng CY. COVID-19 vaccine-associated vitiligo: A cross-sectional study in a tertiary referral center and systematic review. J Dermatol 2023. [PMID: 37186102 DOI: 10.1111/1346-8138.16799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
As the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus continues to infect patients globally, vaccination remains one of the primary methods to combat this prolonged pandemic. However, there are growing reports of coronavirus disease 2019 (COVID-19) vaccines possibly triggering autoimmunity, irrespective of the vaccine's design. This phenomenon has been observed in patients with vitiligo, with a rising number of cases reporting new-onset or worsening vitiligo following COVID-19 vaccinations. In this study, the authors present the most extensive case series of COVID-19 vaccine-associated vitiligo to date, along with a systematic review of the literature. The aim is to assist physicians in the clinical evaluation of patients with vitiligo with regard to future vaccinations.
Collapse
Affiliation(s)
- Tsung-Fu Tsai
- Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan
- Vitiligo Clinic and Pigment Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chau Yee Ng
- Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan
- Vitiligo Clinic and Pigment Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology and Aesthetic Medicine Center, Jen Ai Hospital, Taichung, Taiwan
| |
Collapse
|
42
|
Guo M, Liu X, Chen X, Li Q. Insights into new-onset autoimmune diseases after COVID-19 vaccination. Autoimmun Rev 2023; 22:103340. [PMID: 37075917 PMCID: PMC10108562 DOI: 10.1016/j.autrev.2023.103340] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in more than 670 million infections and almost 7 million deaths globally. The emergence of numerous SARS-CoV-2 has heightened public concern regarding the future course of the epidemic. Currently, the SARS-CoV-2 Omicron variant has rapidly become globally dominant in the COVID-19 pandemic due to its high infectivity and immune evasion. Consequently, vaccination implementation is critically significant. However, growing evidence suggests that COVID-19 vaccination may cause new-onset autoimmune diseases, including autoimmune glomerulonephritis, autoimmune rheumatic diseases, and autoimmune hepatitis. Nevertheless, the causal relationship between COVID-19 vaccines and these autoimmune diseases remains to be demonstrated. In this review, we provide evidence that vaccination induces autoimmunity and summarize possible mechanisms of action, such as molecular mimicry, activation by bystanders, and adjuvants. Our objective is not to refute the importance of vaccines, but to raise awareness about the potential risks of COVID-19 vaccination. In fact, we believe that the benefits of vaccination far outweigh the possible risks and encourage people to get vaccinated.
Collapse
Affiliation(s)
- Ming Guo
- Hebei General Hosptial, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Xiaoxiao Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
43
|
Böröcz K, Kinyó Á, Simon D, Erdő-Bonyár S, Németh P, Berki T. Complexity of the Immune Response Elicited by Different COVID-19 Vaccines, in the Light of Natural Autoantibodies and Immunomodulatory Therapies. Int J Mol Sci 2023; 24:ijms24076439. [PMID: 37047412 PMCID: PMC10094397 DOI: 10.3390/ijms24076439] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Despite the abundance of data on the COVID-19 vaccine-induced immune activation, the impact of natural autoantibodies (nAAbs) on these processes is less well defined. Therefore, we investigated potential connections between vaccine efficacy and nAAb levels. We were also interested in the impact of immunomodulatory therapies on vaccine efficacy. Clinical residual samples were used for the assessment of the COVID-19 vaccine-elicited immune response (IR) (n=255), as well as for the investigation of the immunization-associated expansion of the nAAb pool (n=185). In order to study the potential interaction between immunomodulatory therapies and the vaccine-induced IR, untreated, healthy individuals and patients receiving anti-TNFα or anti-IL-17 therapies were compared (n total =45). In-house ELISAs (anticitrate synthase, anti-HSP60 and-70) and commercial ELISAs (anti-SARS-CoV-2 ELISAs IgG, IgA, NeutraLISA and IFN-γ release assay 'IGRA') were applied. We found significant differences in the IR given to different vaccines. Moreover, nAAb levels showed plasticity in response to anti-COVID-19 immunization. We conclude that our findings may support the theorem about the non-specific beneficial 'side effects' of vaccination, including the broadening of the nAAb repertoire. Considering immunomodulation, we suggest that anti-TNFα and anti-IL17 treatments may interfere negatively with MALT-associated IR, manifested as decreased IgA titers; however, the modest sample numbers of the herein presented model might be a limiting factor of reaching a more comprehensive conclusion.
Collapse
Affiliation(s)
- Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Ágnes Kinyó
- Department of Dermatology, Venereology and Oncodermatology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Szabina Erdő-Bonyár
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| |
Collapse
|
44
|
The Third Dose of BNT162b2 COVID-19 Vaccine Does Not “Boost” Disease Flares and Adverse Events in Patients with Rheumatoid Arthritis. Biomedicines 2023; 11:biomedicines11030687. [PMID: 36979666 PMCID: PMC10045021 DOI: 10.3390/biomedicines11030687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Data on the risk of adverse events (AEs) and disease flares in autoimmune rheumatic diseases (ARDs) after the third dose of COVID-19 vaccine are scarce. The aim of this multicenter, prospective study is to analyze the clinical and immunological safety of BNT162b2 vaccine in a cohort of rheumatoid arthritis (RA) patients followed-up from the first vaccine cycle to the third dose. The vaccine showed an overall good safety profile with no patient reporting serious AEs, and a low percentage of total AEs at both doses (40/78 (51.3%) and 13/47 (27.7%) patients after the second and third dose, respectively (p < 0.002). Flares were observed in 10.3% of patients after the end of the vaccination cycle and 12.8% after the third dose. Being vaccinated for influenza was inversely associated with the onset of AEs after the second dose, at both univariable (p = 0.013) and multivariable analysis (p = 0.027). This result could allow identification of a predictive factor of vaccine tolerance, if confirmed in larger patient populations. A higher disease activity at baseline was not associated with a higher incidence of AEs or disease flares. Effectiveness was excellent after the second dose, with only 1/78 (1.3%) mild breakthrough infection (BI) and worsened after the third dose, with 9/47 (19.2%) BI (p < 0.002), as a probable expression of the higher capacity of the Omicron variants to escape vaccine recognition.
Collapse
|
45
|
Peikert A, Claggett BL, Kim K, Udell JA, Joseph J, Desai AS, Farkouh ME, Hegde SM, Hernandez AF, Bhatt DL, Gaziano JM, Talbot HK, Yancy C, Anand I, Mao L, Cooper LS, Solomon SD, Vardeny O. Association of post-vaccination adverse reactions after influenza vaccine with mortality and cardiopulmonary outcomes in patients with high-risk cardiovascular disease: the INVESTED trial. Eur J Heart Fail 2023; 25:299-310. [PMID: 36335639 DOI: 10.1002/ejhf.2716] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022] Open
Abstract
AIMS Influenza vaccination is associated with reduced cardiopulmonary morbidity and mortality among patients with heart failure or recent myocardial infarction. The immune response to vaccination frequently results in mild adverse reactions (AR), which leads to vaccine hesitancy. This post hoc analysis explored the association between vaccine-related AR and morbidity and mortality in patients with high-risk cardiovascular disease. METHODS AND RESULTS The INVESTED trial randomized 5260 patients with recent heart failure hospitalization or acute myocardial infarction to high-dose trivalent or standard-dose quadrivalent inactivated influenza vaccine. We examined the association between vaccine-related AR and adverse clinical outcomes across both treatment groups in propensity-adjusted models. Among 5210 participants with available information on post-vaccination symptoms, 1968 participants (37.8%) experienced a vaccine-related AR. Compared to those without AR, post-vaccination AR, most commonly injection site pain (60.3%), were associated with lower risk for the composite of all-cause death or cardiopulmonary hospitalization (hazard ratio [HR] 0.83, 95% confidence interval [CI] 0.75-0.92, p < 0.001), cardiopulmonary hospitalizations (HR 0.85 [95% CI 0.76-0.95], p = 0.003), all-cause death (HR 0.77 [95% CI 0.62-0.96], p = 0.02), cardiovascular hospitalizations (HR 0.88 [95% CI 0.78-0.99], p = 0.03) and non-cardiopulmonary hospitalizations (HR 0.80 [95% CI 0.69-0.92], p = 0.003). While mild (76.4%) and moderate (20.6%) AR were most common and together associated with lower risk for the primary outcome (HR 0.81 [95% CI 0.74-0.90], p < 0.001), severe AR (2.9%) were related to increased risk (HR 1.68 [95% CI 1.17-2.42], p = 0.005). CONCLUSION Mild to moderate post-vaccination reactions after influenza vaccine were associated with reduced risk of cardiopulmonary hospitalizations and all-cause mortality in patients with high-risk cardiovascular disease, while severe reactions may indicate increased risk. Mild to moderate AR to influenza vaccination may be a marker of immune response and should not deter future vaccinations.
Collapse
Affiliation(s)
- Alexander Peikert
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian L Claggett
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacob A Udell
- Peter Munk Cardiac Centre, University Health Network and Women's College Hospital, University of Toronto, Toronto, ONT, Canada
| | - Jacob Joseph
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Boston, MA, USA
| | - Akshay S Desai
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael E Farkouh
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ONT, Canada
| | - Sheila M Hegde
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Deepak L Bhatt
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J Michael Gaziano
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Boston, MA, USA
| | - H Keipp Talbot
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Clyde Yancy
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Inder Anand
- Department of Medicine, University of Minnesota, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Lu Mao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Lawton S Cooper
- National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Orly Vardeny
- Department of Medicine, University of Minnesota, Minneapolis VA Health Care System, Minneapolis, MN, USA
| |
Collapse
|
46
|
Molina Rios S, Rojas Martinez R, Estévez Ramirez GM, Medina YF. Systemic lupus erythematosus and antiphospholipid syndrome after COVID-19 vaccination. A case report. Mod Rheumatol Case Rep 2023; 7:43-46. [PMID: 35246682 PMCID: PMC8903504 DOI: 10.1093/mrcr/rxac018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 05/02/2023]
Abstract
Coronavirus disease 2019 (COVID-19) vaccines have some adverse effects, mostly mild. However, by presenting an immunological challenge to the individual, they could infrequently trigger immune-mediated diseases (IMDs). We report the case of a 42-year-old woman, with no previous medical history, who received the first dose of vaccine against COVID-19 and developed inflammatory arthralgias, associated with sudden-onset dyspnoea and hypoxemia. Pulmonary thromboembolism was documented, and the diagnosis of systemic lupus erythematosus (SLE) and secondary antiphospholipid syndrome (APS) was suspected. Autoantibodies were measured confirming this suspicion. After a few days, she presented a massive pericardial effusion with cardiac tamponade that required surgical management. Treatment with azathioprine, hydroxychloroquine, corticosteroids, and anticoagulation was indicated with improvement of all her symptoms. There is controversy regarding the potential of COVID-19 vaccines to induce autoimmunity. Studies addressing the safety of using these vaccines have reported the occurrence of mild local and systemic reactions, most frequently in young adults. So far, there are few reports of patients who have developed autoimmune or autoinflammatory diseases after getting vaccinated with any of the COVID-19 vaccines. To the best of our knowledge, to date, this is one of the first cases of new-onset SLE and secondary APS after COVID-19 vaccination.
Collapse
Affiliation(s)
- Sebastian Molina Rios
- Corresponding author: Sebastian Molina Rios, Rheumatology Unit. Department of Internal Medicine. School of Medicine, Universidad Nacional de Colombia. Carrera 30 No. 45-03. 111321. Bogotá DC, Colombia. Phone Number: +573213045304, E-mail:
| | - Rigoberto Rojas Martinez
- Rheumatology Unit. Department of Internal Medicine. School of Medicine, Universidad Nacional de Colombia. Carrera 30 No. 45-03. 111321. Bogotá DC, Colombia
- Hospital Universitario Nacional de Colombia. Calle 44 No. 59-75. 111321. Bogotá DC, Colombia
| | - Gloria Marcela Estévez Ramirez
- Rheumatology Unit. Department of Internal Medicine. School of Medicine, Universidad Nacional de Colombia. Carrera 30 No. 45-03. 111321. Bogotá DC, Colombia
- Hospital Universitario Nacional de Colombia. Calle 44 No. 59-75. 111321. Bogotá DC, Colombia
| | - Yimy Francisco Medina
- Rheumatology Unit. Department of Internal Medicine. School of Medicine, Universidad Nacional de Colombia. Carrera 30 No. 45-03. 111321. Bogotá DC, Colombia
- Hospital Universitario Nacional de Colombia. Calle 44 No. 59-75. 111321. Bogotá DC, Colombia
| |
Collapse
|
47
|
Nishimura N, Shiomichi Y, Takeuchi S, Akamine S, Yoneda R, Yoshizawa S. IgA vasculitis following COVID-19 vaccination. Mod Rheumatol Case Rep 2023; 7:122-126. [PMID: 35253880 PMCID: PMC8903512 DOI: 10.1093/mrcr/rxac014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/24/2021] [Accepted: 02/11/2022] [Indexed: 01/07/2023]
Abstract
Immunoglobulin A (IgA) vasculitis is generally triggered by infectious causes, but it has also been reported after immunisation with various vaccines. Herein, we report two cases of IgA vasculitis after receiving the first or second dose of the Pfizer-BioNTech BNT16B2b2 mRNA vaccine. Two men, aged 22 and 30 years, developed palpable purpura on the extremities and arthritis. One patient also complained of fever and gastrointestinal symptoms. Laboratory findings revealed mild leucocytosis and slightly elevated C-reactive protein levels, although the platelet count and coagulation profile were within normal levels in both cases. Proteinuria and microhaematuria were seen in one patient. Skin biopsies were performed in both patients and revealed leucocytoclastic vasculitis. The deposits of IgA and C3 were shown in immunofluorescence studies in one patient. Both patients were diagnosed with IgA vasculitis and treated with prednisolone, and their symptoms resolved within 1 week after initiation of treatment. The coronavirus disease 2019 mRNA vaccine could trigger IgA vasculitis; however, a coincidence cannot be ruled out.
Collapse
Affiliation(s)
- Naoya Nishimura
- Department of Rheumatology, Hamanomachi Hospital, Fukuoka, Japan
| | | | | | - Shun Akamine
- Department of Pathology, Hamanomachi Hospital, Fukuoka, Japan
| | - Reiko Yoneda
- Department of Pathology, Hamanomachi Hospital, Fukuoka, Japan
| | - Seiji Yoshizawa
- Department of Rheumatology, Hamanomachi Hospital, Fukuoka, Japan
| |
Collapse
|
48
|
dos Santos-Silva E, Torres-Rêgo M, Gláucia-Silva F, Feitosa RC, Lacerda AF, Rocha HADO, Fernandes-Pedrosa MDF, da Silva-Júnior AA. Cationic PLGA Nanoparticle Formulations as Biocompatible Immunoadjuvant for Serum Production and Immune Response against Bothrops jararaca Venom. Toxins (Basel) 2022; 14:toxins14120888. [PMID: 36548785 PMCID: PMC9786128 DOI: 10.3390/toxins14120888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Snakebite envenoming represents a worldwide public health issue. Suitable technologies have been investigated for encapsulated recombinant or native proteins capable of inducing an effective and long-lasting adaptive immune response. Nanoparticles are colloidal dispersions that have been used as drug delivery systems for bioactive biological compounds. Venom-loaded nanoparticles modulate the protein release and activate the immune response to produce specific antibodies. In this study, biocompatible cationic nanoparticles with Bothrops jararaca venom were prepared to be used as a novel immunoadjuvant that shows a similar or improved immune response in antibody production when compared to a conventional immunoadjuvant (aluminum hydroxide). We prepared stable, small-sized and spherical particles with high Bothrops jararaca venom protein association efficiency. The high protein loading efficiency, electrophoresis, and zeta potential results demonstrated that Bothrops jararaca venom is adsorbed on the particle surface, which remained as a stable colloidal dispersion over 6 weeks. The slow protein release occurred and followed parabolic diffusion release kinetics. The in vivo studies demonstrated that venom-loaded nanoparticles were able to produce an immune response similar to that of aluminum hydroxide. The cationic nanoparticles (CNp) as carriers of bioactive molecules, were successfully developed and demonstrated to be a promising immunoadjuvant.
Collapse
Affiliation(s)
- Emanuell dos Santos-Silva
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | - Manoela Torres-Rêgo
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | - Fiamma Gláucia-Silva
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | - Renata Carvalho Feitosa
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | - Ariane Ferreira Lacerda
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | | | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte (UFRN), Natal-RN 59072-970, Brazil
- Correspondence:
| |
Collapse
|
49
|
Immune-Mediated Diseases Following COVID-19 Vaccination: Report of a Teaching Hospital-Based Case-Series. J Clin Med 2022; 11:jcm11247484. [PMID: 36556100 PMCID: PMC9785620 DOI: 10.3390/jcm11247484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The occurrence and course of immune-mediated diseases (IMDs) following COVID-19 vaccination has been little explored so far. We retrieved, among adult patients hospitalized at the Internal Department of a French university hospital up to May 2022, all those who had developed, or relapsed to, an IMD less than 3 weeks following COVID-19 vaccination, without other triggers. Twenty-seven (24 new-onset) post-COVID-19 vaccine IMDs were recorded. They comprised giant cell arteritis or polymyalgia rheumatica (n = 16, HLA-DRB1*04 in 58% of 12 assessed GCA cases), immune-mediated necrotizing myositis or acute rhabdomyolysis, systemic vasculitis, immune thrombocytopenic purpura, rheumatoid arthritis, anti-synthetase syndrome, and adult-onset Still's disease. The causative vaccines were mRNA-based (20 cases) or viral vector-based (7 cases). The IMD typically occurred after the first vaccine dose, with an average delay of 8 (5 SD) days. The patients' mean age was 67 years, and 58% were women. The IMDs had protracted courses in all but three of the patients and typically required high-dose glucocorticoids, in combination with immunomodulators in 13 patients. One patient died of intractable rhabdomyolysis, whereas five suffered permanent damage from IMDs. Eleven patients with well-controlled IMDs completed their COVID-19 vaccination schedule, and two suffered mild IMD relapses. There is a risk of IMDs, notably GCA/PMR, and muscle disorders, following COVID-19 vaccination. Such adverse reactions typically occurred after the first dose, raising concern about subsequent COVID-19 vaccinations. However, early re-challenge in well-controlled IMDs appeared safe.
Collapse
|
50
|
Toward Establishing an Ideal Adjuvant for Non-Inflammatory Immune Enhancement. Cells 2022; 11:cells11244006. [PMID: 36552770 PMCID: PMC9777512 DOI: 10.3390/cells11244006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The vertebrate immune system functions to eliminate invading foreign nucleic acids and foreign proteins from infectious diseases and malignant tumors. Because pathogens and cancer cells have unique amino acid sequences and motifs (e.g., microbe-associated molecular patterns, MAMPs) that are recognized as "non-self" to the host, immune enhancement is one strategy to eliminate invading cells. MAMPs contain nucleic acids specific or characteristic of the microbe and are potential candidates for immunostimulants or adjuvants. Adjuvants are included in many vaccines and are a way to boost immunity by deliberately administering them along with antigens. Although adjuvants are an important component of vaccines, it is difficult to evaluate their efficacy ex vivo and in vivo on their own (without antigens). In addition, inflammation induced by currently candidate adjuvants may cause adverse events, which is a hurdle to their approval as drugs. In addition, the lack of guidelines for evaluating the safety and efficacy of adjuvants in drug discovery research also makes regulatory approval difficult. Viral double-stranded (ds) RNA mimics have been reported as potent adjuvants, but the safety barrier remains unresolved. Here we present ARNAX, a noninflammatory nucleic acid adjuvant that selectively targets Toll-like receptor 3 (TLR3) in antigen-presenting dendritic cells (APCs) to safely induce antigen cross-presentation and subsequently induce an acquired immune response independent of inflammation. This review discusses the challenges faced in the clinical development of novel adjuvants.
Collapse
|