51
|
Recent advances in treatment Crimean-Congo hemorrhagic fever virus: A concise overview. Microb Pathog 2022; 169:105657. [PMID: 35753597 DOI: 10.1016/j.micpath.2022.105657] [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: 02/20/2022] [Revised: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
The Crimean Congo Hemorrhagic Fever Virus (CCHFV) is widespread in Africa, Asia, and Europe, among other places. The disease was initially discovered in the Crimean cities of the Soviet Union and the Congo, and it was given the name Crimean Congo because it induces hemorrhagic fever. According to studies, when the virus enters the body, it settles in immune cells such as macrophages and dendritic cells, causing them to malfunction and secrete inflammatory cytokines such as TNF-alpha, IL1, and IL6, resulting in cytokine storms that induces shock via endothelial activation and vascular leakage, while on the other hand, clots and disseminated intravascular coagulation (DIC) formation causes massive defects in various organs such as the liver and kidneys, as well as fatal bleeding. Disease prevention and treatment are crucial since no other effective vaccination against the disease has yet been developed. Immunotherapy is utilized as a consequence. One of the most effective treatments, when combined with compensatory therapies such as blood and platelet replacement, water, electrolytes, Fresh Frozen Plasma (FFP) replacement, and other compensatory therapies, is one of the most effective treatments. Studies; show that immunotherapy using IVIG and neutralizing and non-neutralizing monoclonal antibodies; cytokine therapy, and anti-inflammatory therapy using corticosteroids are effective ways to treat the disease.
Collapse
|
52
|
Kwon B, Yang SJ, Cho SM, Kim ME, Nahm DH. Intramuscular administration of autologous total immunoglobulin G induces immunomodulatory effects on T cells in healthy human subjects: An open-labeled prospective single-arm trial. Medicine (Baltimore) 2022; 101:e29486. [PMID: 35665739 PMCID: PMC9276166 DOI: 10.1097/md.0000000000029486] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND We hypothesized that intramuscular administration of autologous total immunoglobulin G (IgG) could induce an immunomodulatory effect in human subjects. In our previous studies, we showed that intramuscular administration of autologous total IgG could induce significant clinical improvements and increases of the serum levels of interleukin-10 (IL-10) and interferon-gamma (IFN-γ) in patients with atopic dermatitis. OBJECTIVE To investigate the mechanism of immunomodulation induced by intramuscular administration of autologous total IgG, we evaluated changes in T cells before and after intramuscular administrations of autologous total IgG in this study. METHODS Thirteen healthy adults received 8 intramuscular injections of 50 mg autologous total IgG for 4 weeks (from week 0 to week 4). The percentages of IL-10- or IFN-γ-producing peripheral blood T cells, as well as serum levels of IL-10, IFN-γ, and immunoglobulins, were measured at baseline (week 0) and at weeks 4, 8, and 12. RESULTS The percentage of IL-10-producing CD4+ T cells was significantly increased at weeks 8 and 12 compared to baseline (P < .05), while the percentage of IFN-γ-producing CD3+ T cells was significantly increased at week 12 compared to baseline (P < .05). There were no significant differences in the serum levels of IL-10, IFN-γ, and immunoglobulins before and after intramuscular administration of autologous total IgG (P > .05). No serious adverse events were observed. CONCLUSION Intramuscular administration of autologous total IgG induced immunomodulatory effects on T cells in healthy human subjects. This simple intervention could be a safe, effective, and economical T cell immunomodulation method for human subjects (NCT03695757).
Collapse
|
53
|
Gao Y, Jin H. Efficacy and safety of intravenous immunoglobulin for treating refractory livedoid vasculopathy: a systematic review. Ther Adv Chronic Dis 2022; 13:20406223221097331. [PMID: 35634570 PMCID: PMC9134453 DOI: 10.1177/20406223221097331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/12/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction: Intravenous immunoglobulin (IVIG) was reported to be the third most used
monotherapy in livedoid vasculopathy (LV). There is currently a lack of
randomized controlled clinical trials and no standardized therapeutic
regimen for IVIG therapy in LV. Methods: We performed a systematic review of the efficacy and safety of IVIG in
treating patients with LV using PubMed, Cochrane, and Embase databases. Results: Eighty LV patients from 17 articles were included, receiving IVIG therapy at
a dose of 1–2.1 g/kg body weight every 4 weeks. The effective rate of IVIG
therapy in LV patients was 95% (76/80) in published studies, showing a good
clinical response for resolution of pain, skin ulcerations, and neurological
symptoms, and reducing the dependence on glucocorticoids and
immunosuppressive agents. IVIG therapy was well tolerated, and no severe
adverse events were observed. Conclusion: Overall, to a certain degree, IVIG is probably a safe and effective treatment
alternative for refractory LV patients, which still need to be confirmed by
large-scale randomized controlled clinical trials.
Collapse
Affiliation(s)
- Yimeng Gao
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Hongzhong Jin
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, No.1 Shuaifuyuan, Dongcheng district, Beijing, 100730 China
| |
Collapse
|
54
|
Branch DR, Almizraq RJ, Bussel JB. Hemolysis after high-dose intravenous immunoglobulin: An under-appreciated sequelae. Am J Hematol 2022; 97:E183-E185. [PMID: 35170068 DOI: 10.1002/ajh.26501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Donald R. Branch
- Centre for Innovation Canadian Blood Services Toronto Ontario Canada
- Department of Medicine University of Toronto Toronto Ontario Canada
| | | | - James B. Bussel
- Department of Pediatrics Weill Cornell Medical College New York New York USA
| |
Collapse
|
55
|
Abstract
Neuroimmunological diseases and their treatment compromise the immune system, thereby increasing the risk of infections and serious illness. Consequently, vaccinations to protect against infections are an important part of the clinical management of these diseases. However, the wide variety of immunotherapies that are currently used to treat neuroimmunological disease — particularly multiple sclerosis and neuromyelitis optica spectrum disorders — can also impair immunological responses to vaccinations. In this Review, we discuss what is known about the effects of various immunotherapies on immunological responses to vaccines and what these effects mean for the safe and effective use of vaccines in patients with a neuroimmunological disease. The success of vaccination in patients receiving immunotherapy largely depends on the specific mode of action of the immunotherapy. To minimize the risk of infection when using immunotherapy, assessment of immune status and exclusion of underlying chronic infections before initiation of therapy are essential. Selection of the required vaccinations and leaving appropriate time intervals between vaccination and administration of immunotherapy can help to safeguard patients. We also discuss the rapidly evolving knowledge of how immunotherapies affect responses to SARS-CoV-2 vaccines and how these effects should influence the management of patients on these therapies during the COVID-19 pandemic. In this Review, the authors discuss how various immunotherapies for neuroimmunological diseases interact with vaccination responses, including responses to SARS-CoV-2 vaccinations, and the implications for the safe and effective use of vaccines in patients with these diseases. Vaccination against infection is an essential part of the management of neuroimmunological diseases. All indicated vaccinations should be administered before initiation of immunotherapy whenever possible; appropriate intervals between vaccination and treatment vary with treatment and vaccination. Inactivated vaccines are considered safe in neuroimmunological diseases but live vaccines are generally contraindicated during immunotherapy. Vaccination responses during immunotherapy can be diminished or abrogated, depending on the treatment and vaccination; antibody titre testing to monitor responses can be considered where appropriate. Vaccinations must be avoided during relapses or exacerbations of neuroimmunological diseases. Vaccination against SARS-CoV-2 is recommended for patients with neuroimmunological disease but some immunotherapies limit the immune response; therefore, timing should be considered carefully.
Collapse
|
56
|
Takeuchi E, Kajiyama Y, Ando K, Funaki S, Okuno T, Shintani Y, Mochizuki H. [The efficacy of eculizumab against post-thymectomy exacerbations in thymoma associated myasthenia gravis (MG)]. Rinsho Shinkeigaku 2022; 62:277-280. [PMID: 35354724 DOI: 10.5692/clinicalneurol.cn-001682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We report a 62-year-old woman with thymoma associated myasthenia gravis (MG). She had significant dysphagia and was treated with corticosteroids, intravenous immunoglobulin (IVIG), immunoadsorption plasmapheresis (IAPP), and immunosuppressive drugs, and the extended thymectomy. Her symptoms gradually improved, but 3 weeks after thymectomy, her bulbar symptoms recurred. Although she was treated with repeated IVIG and IAPP, her symptom remained. Finally, after starting eculizumab did her symptoms go into complete remission. This case suggests the efficacy of anti-complement therapy for postoperative exacerbation of MG.
Collapse
Affiliation(s)
- Eriko Takeuchi
- Department of Neurology, Osaka University Graduate School of Medicine
| | - Yuta Kajiyama
- Department of Neurology, Osaka University Graduate School of Medicine.,Department of Neurology, Sakai City Medical Center
| | - Koshiro Ando
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine.,Department of General Thoracic Surgery, Osaka Habikino Medical Center
| | - Soichiro Funaki
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine
| |
Collapse
|
57
|
Lee ST, Lee HS, Lee WJ, Cha HA, Kim SH, Shin SY, Chu K, Lee SK. The safety and efficacy of intravenous immunoglobulin in autoimmune encephalitis. Ann Clin Transl Neurol 2022; 9:610-621. [PMID: 35315247 PMCID: PMC9082384 DOI: 10.1002/acn3.51540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 02/25/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Although intravenous immunoglobulin (IVIG) is the first-line immunotherapy in autoimmune encephalitis, all supporting evidence comes from retrospective case series. Here, we performed a prospective clinical trial of IVIG for functional recovery in autoimmune encephalitis. METHODS This single-arm, open-label study assessed the efficacy and safety of 10% intravenous IVIG treatment in newly diagnosed patients with possible autoimmune encephalitis. Patients received IVIG (0.4 g/kg/day) for 5 days. Rescue immunotherapy was permitted when the patient deteriorated before day 8 or showed no improvement at day 8. The primary outcome was the change in the modified Rankin Scale (mRS) score at day 8 and 29. The secondary outcomes were the mRS score improvement and the score changes and improvements on four other clinical scales. RESULTS Overall, 23 patients received IVIG (intension-to-treat, ITT), and 18 patients completed the study according to the protocol (per-protocol, PP). mRS improved significantly at days 8 and 29 compared to baseline in both the ITT and PP populations. Other secondary outcomes also improved significantly at day 8, 15, and 29 versus baseline. In the PP population, 6/18 patients achieved favorable outcomes with IVIG alone (mRS = 0~2 at day 8), and 12/18 patients received rescue immunotherapy. Five adverse events were reported in relation to IVIG, all of which were mild. INTERPRETATION IVIG improved neurological functional outcomes, and the improvement was evident by day 8. Adverse effects were tolerable. These data provide the prospective evidence regarding the efficacy of IVIG in improving the functional outcomes of autoimmune encephalitis.
Collapse
Affiliation(s)
- Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Han Sang Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Woo-Jin Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Han-A Cha
- GC Pharma, Yongin-si, Gyeonggi-do, South Korea
| | | | - Seo-Yi Shin
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Kon Chu
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
58
|
Habets DHJ, Pelzner K, Wieten L, Spaanderman MEA, Villamor E, Al-Nasiry S. Intravenous immunoglobulins improve live birth rate among women with underlying immune conditions and recurrent pregnancy loss: a systematic review and meta-analysis. Allergy Asthma Clin Immunol 2022; 18:23. [PMID: 35277202 PMCID: PMC8917719 DOI: 10.1186/s13223-022-00660-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Intravenous immunoglobulin (IVIG) is increasingly used as a treatment for recurrent pregnancy loss (RPL) despite lack of clear evidence on efficacy. Recent data suggest IVIG might be more effective in a subgroup of women with an aberrant immunological profile. Therefore, a systematic review and meta-analysis of studies on the effectiveness of IVIG treatment on pregnancy outcome among women with RPL and underlying immunological conditions (e.g., elevated NK cell percentage, elevated Th1/Th2 ratio, diagnosis with autoimmune disorders) was conducted. Eight non-randomized controlled trials, including 478 women (intervention: 284; control: 194), met eligibility criteria. Meta-analysis showed that treatment with IVIG was associated with a two-fold increase in live birth rate (RR 1.98, 95% CI 1.44–2.73, P < 0.0001). The effect of IVIG was particularly marked in the subgroup of studies including patients based on presence of elevated (> 12%) NK-cell percentage (RR 2.32, 95% CI 1.77–3.02, P < 0.0001) and when starting intervention prior to or during cycle of conception (RR 4.47, 95% CI 1.53–13.05, P = 0.006). In conclusion, treatment with IVIG may improve live birth rate in women with RPL and underlying immune conditions. However, these results should be interpreted with caution as studies are limited by low number of participants and the non-randomized design, which represent seriously biases. Future randomized controlled trials in women with RPL and underlying immune conditions are needed before using IVIG in a clinical setting.
Collapse
Affiliation(s)
- Denise H J Habets
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands. .,Department of Transplantation Immunology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands. .,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| | - Kim Pelzner
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Marc E A Spaanderman
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Eduardo Villamor
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Salwan Al-Nasiry
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
59
|
Rodriguez A, Klein CJ, Sechi E, Alden E, Basso MR, Pudumjee S, Pittock SJ, McKeon A, Britton JW, Lopez-Chiriboga AS, Zekeridou A, Zalewski NL, Boeve BF, Day GS, Gadoth A, Burkholder D, Toledano M, Dubey D, Flanagan EP. LGI1 antibody encephalitis: acute treatment comparisons and outcome. J Neurol Neurosurg Psychiatry 2022; 93:309-315. [PMID: 34824144 PMCID: PMC8862031 DOI: 10.1136/jnnp-2021-327302] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To compare acute treatment responses and long-term outcome in leucine-rich glioma-inactivated 1 (LGI1) antibody encephalitis. METHODS Retrospective case series of 118 patients with LGI1 antibody encephalitis evaluated at Mayo Clinic across all US sites from 1 May 2008 to 31 March 2019. Patient clinical data were identified and analysed through the neuroimmunology laboratory and electronic medical record. LGI1 antibody detection was by cell-based indirect immunofluorescence assay of serum, cerebrospinal fluid or both. Clinical outcomes were faciobrachial dystonic seizure (FBDS) resolution, modified Rankin Scale (mRS) score, Kokmen Short Test of Mental Status (STMS) score (0-38 point scale) and neuropsychometric testing results. RESULTS Compared with intravenous immunoglobulin (IVIg) (n=21), patients treated with single-agent acute corticosteroids (intravenous, oral or both) (n=49) were more likely to experience resolution of FBDS (61% vs 7%, p=0.002) and improvements in mRS score (ΔmRS score 2 vs 0, p=0.008) and median Kokmen STMS scores (ΔKokmen STMS score 5 points vs 0 points, p=0.01). In 54 patients with long-term follow-up (≥2 years), the median mRS score was 1 (range 0-6) and the median Kokmen STMS score was 36 (range 24-38) after all combinations of immunotherapy. Neuropsychometric testing in 32 patients with long-term follow-up (≥2 years) demonstrated short-term memory impairments in 37%. CONCLUSIONS Corticosteroids appeared more effective acutely than IVIg in improving LGI1 antibody encephalitis in this retrospective comparison of immunotherapies. While improvement with immunotherapy is typical and long-term outcome is favourable, short-term memory deficits are noted in approximately a third of the patients.
Collapse
Affiliation(s)
| | - C J Klein
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elia Sechi
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eva Alden
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael R Basso
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shehroo Pudumjee
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sean J Pittock
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew McKeon
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Anastasia Zekeridou
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - B F Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Hospital Jacksonville, Jacksonville, Florida, USA
| | - Avi Gadoth
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Michel Toledano
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Divyanshu Dubey
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eoin P Flanagan
- Department of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
60
|
Chen DTL, Chang JPC, Cheng SW, Chang HC, Hsu JH, Chang HH, Chiu WC, Su KP. Kawasaki disease in childhood and psychiatric disorders: A population-based case-control prospective study in Taiwan. Brain Behav Immun 2022; 100:105-111. [PMID: 34848339 DOI: 10.1016/j.bbi.2021.11.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Kawasaki disease (KD) is a common childhood acute inflammatory disease and potentially triggers a chronic inflammation. Although some researches have investigated neurodevelopmental consequences following KD, the findings have been inconsistent. This is the first population-based study targeted on KD and common psychiatric disorders. OBJECTIVES We aimed to investigate the association between KD and psychiatric disorders and hypothesized that standard anti-inflammatory treatment by intravenous immunoglobulin (IVIG) may protect against development of psychiatric disorders. METHOD We retrieved data from Taiwan's National Health Insurance Research database (NHIRD). Patients (n = 282,513) with psychiatric disorders (the case group) during 1997-2013 were included, and the control group was matched with age, sex, income and urbanization (1:1). We calculated the prevalence of KD in both groups and estimated odd ratios (ORs) and 95% confidence intervals (CIs) in the subgroup analyses for KD in conditions of age, severity, and common psychiatric comorbidity. RESULTS Numbers of patients with KD were 460 in the cases and 380 in the controls (p = .006), and the crude OR of KD was 1.21 times greater (95% CI = 1.06-1.39, p = .006) in the case than the control groups. KD patients without IVIG treatment (n = 126) were higher in the cases than those in the controls (n = 54), with the OR of 2.33 (95% CI = 1.70-3.21, p < .0001). Subgroup analyses showed that KD survivors were at significant risk for autism spectrum disorders (ASD) (OR = 2.15, 95% CI = 1.27-3.65; p = .005) and attention deficit and hyperactivity disorders (ADHD) (OR = 1.19, 95% CI = 1.02-1.39; p = 0.03), and a trend of increased risk for anxiety disorders (OR = 1.36, 95%CI = 0.99-1.86; p = 0.05). CONCLUSIONS Patients with KD were more likely to have comorbid psychiatric disorders, including ASD and ADHD. Moreover, anti-inflammatory treatment with IVIG may have potential prophylactic effects against the development of psychiatric disorders.
Collapse
Affiliation(s)
- Daniel Tzu-Li Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Biomedicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Jane Pei-Chen Chang
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Biomedicine, College of Medicine, China Medical University, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Szu-Wei Cheng
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Chih Chang
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, and Chinese Medicine Research Center, China Medical University, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Che Chiu
- School of Medicine, Fu Jen Catholic University, Taipei, Taiwan; Department of Psychiatry, Cathay General Hospital, Taipei, Taiwan.
| | - Kuan-Pin Su
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Biomedicine, College of Medicine, China Medical University, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| |
Collapse
|
61
|
Meenan J, Hall R, Badle S, Chatterjee B, Win N, Tsitsikas DA. Tocilizumab in the management of posttransfusion hyperhemolysis syndrome in sickle cell disease: The experience so far. Transfusion 2022; 62:546-550. [PMID: 35092617 DOI: 10.1111/trf.16805] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Posttransfusion hyperhemolysis syndrome is a rare but life-threatening form of delayed hemolytic transfusion reaction with lysis of both transfused and autologous red cells, seen predominantly in patients with sickle cell disease. Macrophage activation is thought to play a major role in its pathophysiology. Standard treatment is with intravenous immunoglobulin and steroids but refractory cases pose a major clinical problem. Tocilizumab is a humanized monoclonal antibody against the IL-6 receptor that can inhibit IL-6 induced macrophage activation. METHODS AND MATERIALS We describe the case of a 33-year-old woman with sickle cell anemia and posttransfusion hyper hemolysis syndrome refractory to standard therapy, treated with Tocilizumab. We also review all cases reported in the literature where Tocilizumab was used for posttransfusion hyperhemolysis. RESULTS Treatment with Tocilizumab was well tolerated with no observed adverse events. There was no further drop in Hb after day 2 of treatment with subsequent continuous gradual improvement. Her bilirubin dropped significantly after the first dose and continued to improve, while ferritin and LDH reduced significantly after day 2 of treatment with Tocilizumab and continued to drop thereafter. Like in our case, all other cases in the literature where Tocilizumab was used for posttransfusion hyperhemolysis led to rapid clinical responses and no adverse events. DISCUSSION Even though the number of cases of posttransfusion hyper hemolysis syndrome treated with Tocilizumab are few, they have all been associated with rapid clinical responses with no observed adverse events suggesting that the role of Tocilizumab in this context needs to be further explored.
Collapse
Affiliation(s)
- John Meenan
- Haemoglobinopathy Service, Department of Haematology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Rhys Hall
- Haemoglobinopathy Service, Department of Haematology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Saket Badle
- Haemoglobinopathy Service, Department of Haematology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Basabi Chatterjee
- Haemoglobinopathy Service, Department of Haematology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Nay Win
- National Health Service Blood and Transplant, Tooting Centre, London, UK
| | - Dimitris A Tsitsikas
- Haemoglobinopathy Service, Department of Haematology, Homerton University Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
62
|
Mimura Y, Mimura-Kimura Y, Saldova R, Rudd PM, Jefferis R. Enhanced Immunomodulatory Effect of Intravenous Immunoglobulin by Fc Galactosylation and Nonfucosylation. Front Immunol 2022; 13:818382. [PMID: 35154135 PMCID: PMC8831331 DOI: 10.3389/fimmu.2022.818382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is used as an immunomodulatory agent in the treatment of various autoimmune/inflammatory diseases although its mechanism of action remains elusive. Recently, nonfucosylated IgG has been shown to be preferentially bound to Fcγ receptor IIIa (FcγRIIIa) on circulating natural killer cells; therefore, we hypothesized that nonfucosylated IVIG may modulate immune responses through FcγRIIIa blockade. Here, homogeneous fucosylated or nonfucosylated glycoforms of normal polyclonal IgG bearing sialylated, galactosylated or nongalactosylated Fc oligosaccharides were generated by chemoenzymatic glycoengineering to investigate whether the IgG glycoforms can inhibit antibody-dependent cellular cytotoxicity (ADCC). Among the six IgG glycoforms, galactosylated, nonfucosylated IgG [(G2)2] had the highest affinity to FcγRIIIa and 20 times higher potency to inhibit ADCC than native IgG. A pilot study of IVIG treatment in mice with collagen antibody-induced arthritis highlighted the low-dose (G2)2 glycoform of IVIG (0.1 g/kg) as an effective immunomodulatory agent as the 10-fold higher dose of native IVIG. These preliminary results suggest that the anti-inflammatory activity of IVIG is in part mediated via activating FcγR blockade by galactosylated, nonfucosylated IgG and that such nonfucosylated IgG glycoforms bound to FcγRs on immune cells play immunomodulatory roles in health and disease. This study provides insights into improved therapeutic strategies for autoimmune/inflammatory diseases using glycoengineered IVIG and recombinant Fc.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
- *Correspondence: Yusuke Mimura,
| | - Yuka Mimura-Kimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Dublin, Ireland
| | - Pauline M. Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Dublin, Ireland
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Centros, Singapore
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
63
|
Alijotas-Reig J, Esteve-Valverde E, Anunciación-Llunell A, Marques-Soares J, Pardos-Gea J, Miró-Mur F. Pathogenesis, Diagnosis and Management of Obstetric Antiphospholipid Syndrome: A Comprehensive Review. J Clin Med 2022; 11:675. [PMID: 35160128 PMCID: PMC8836886 DOI: 10.3390/jcm11030675] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Antiphospholipid syndrome is an autoimmune disorder characterized by vascular thrombosis and/or pregnancy morbidity associated with persistent antiphospholipid antibody positivity. Cases fulfilling the Sydney criteria for obstetric morbidity with no previous thrombosis are known as obstetric antiphospholipid syndrome (OAPS). OAPS is the most identified cause of recurrent pregnancy loss and late-pregnancy morbidity related to placental injury. Cases with incomplete clinical or laboratory data are classified as obstetric morbidity APS (OMAPS) and non-criteria OAPS (NC-OAPS), respectively. Inflammatory and thrombotic mechanisms are involved in the pathophysiology of OAPS. Trophoblasts, endothelium, platelets and innate immune cells are key cellular players. Complement activation plays a crucial pathogenic role. Secondary placental thrombosis appears by clot formation in response to tissue factor activation. New risk assessment tools could improve the prediction of obstetric complication recurrences or thromboses. The standard-of-care treatment consists of low-dose aspirin and prophylactic low molecular weight heparin. In refractory cases, the addition of hydroxychloroquine, low-dose prednisone or IVIG improve pregnancy outcomes. Statins and eculizumab are currently being tested for treating selected OAPS women. Finally, we revisited recent insights and concerns about the pathophysiology, diagnosis and management of OAPS.
Collapse
Affiliation(s)
- Jaume Alijotas-Reig
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Enrique Esteve-Valverde
- Department of Internal Medicine, Althaia Xarxa Assistencial, Carrer Dr Joan Soler 1-3, 08243 Manresa, Spain;
| | - Ariadna Anunciación-Llunell
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
| | - Joana Marques-Soares
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Josep Pardos-Gea
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron Hospital Campus, Hospital Universitari Vall d’Hebron (HUVH), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Francesc Miró-Mur
- Systemic Autoimmune Diseases Research Unit, Vall d’Hebron Hospital Campus, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.A.-L.); (J.M.-S.); (J.P.-G.)
| |
Collapse
|
64
|
Benfaremo D, Svegliati S, Paolini C, Agarbati S, Moroncini G. Systemic Sclerosis: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2022; 10:biomedicines10010163. [PMID: 35052842 PMCID: PMC8773282 DOI: 10.3390/biomedicines10010163] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Systemic sclerosis (SSc) is a systemic, immune-mediated chronic disorder characterized by small vessel alterations and progressive fibrosis of the skin and internal organs. The combination of a predisposing genetic background and triggering factors that causes a persistent activation of immune system at microvascular and tissue level is thought to be the pathogenetic driver of SSc. Endothelial alterations with subsequent myofibroblast activation, excessive extracellular matrix (ECM) deposition, and unrestrained tissue fibrosis are the pathogenetic steps responsible for the clinical manifestations of this disease, which can be highly heterogeneous according to the different entity of each pathogenic step in individual subjects. Although substantial progress has been made in the management of SSc in recent years, disease-modifying therapies are still lacking. Several molecular pathways involved in SSc pathogenesis are currently under evaluation as possible therapeutic targets in clinical trials. These include drugs targeting fibrotic and metabolic pathways (e.g., TGF-β, autotaxin/LPA, melanocortin, and mTOR), as well as molecules and cells involved in the persistent activation of the immune system (e.g., IL4/IL13, IL23, JAK/STAT, B cells, and plasma cells). In this review, we provide an overview of the most promising therapeutic targets that could improve the future clinical management of SSc.
Collapse
Affiliation(s)
- Devis Benfaremo
- Clinica Medica, Department of Internal Medicine, Ospedali Riuniti “Umberto I-G.M. Lancisi-G. Salesi”, 60126 Ancona, Italy;
| | - Silvia Svegliati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Chiara Paolini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Silvia Agarbati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Internal Medicine, Ospedali Riuniti “Umberto I-G.M. Lancisi-G. Salesi”, 60126 Ancona, Italy;
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
- Correspondence:
| |
Collapse
|
65
|
Nano Differential Scanning Fluorimetry-Based Thermal Stability Screening and Optimal Buffer Selection for Immunoglobulin G. Pharmaceuticals (Basel) 2021; 15:ph15010029. [PMID: 35056086 PMCID: PMC8778976 DOI: 10.3390/ph15010029] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Nano differential scanning fluorimetry (nanoDSF) is a high-throughput protein stability screening technique that simultaneously monitors protein unfolding and aggregation properties. The thermal stability of immunoglobulin G (IgG) was investigated in three different buffers (sodium acetate, sodium citrate, and sodium phosphate) ranging from pH 4 to 8. In all three buffers, the midpoint temperature of thermal unfolding (Tm) showed a tendency to increase as the pH increased, but the aggregation propensity was different depending on the buffer species. The best stability against aggregation was obtained in the sodium acetate buffers below pH 4.6. On the other hand, IgG in the sodium citrate buffer had higher aggregation and viscosity than in the sodium acetate buffer at the same pH. Difference of aggregation between acetate and citrate buffers at the same pH could be explained by a protein-protein interaction study, performed with dynamic light scattering, which suggested that intermolecular interaction is attractive in citrate buffer but repulsive in acetate buffer. In conclusion, this study indicates that the sodium acetate buffer at pH 4.6 is suitable for IgG formulation, and the nanoDSF method is a powerful tool for thermal stability screening and optimal buffer selection in antibody formulations.
Collapse
|
66
|
Bohländer F, Weißmüller S, Riehl D, Gutscher M, Schüttrumpf J, Faust S. The Functional Role of IgA in the IgM/IgA-Enriched Immunoglobulin Preparation Trimodulin. Biomedicines 2021; 9:1828. [PMID: 34944644 PMCID: PMC8698729 DOI: 10.3390/biomedicines9121828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
In comparison to human immunoglobulin (Ig) G, antibodies of IgA class are not well investigated. In line with this, the functional role of the IgA component in IgM/IgA-enriched immunoglobulin preparations is also largely unknown. In recent years, powerful anti-pathogenic and immunomodulatory properties of human serum IgA especially on neutrophil function were unraveled. Therefore, the aim of our work is to investigate functional aspects of the trimodulin IgA component, a new plasma-derived polyvalent immunoglobulin preparation containing ~56% IgG, ~23% IgM and ~21% IgA. The functional role of IgA was investigated by analyzing the interaction of IgA with FcαRI, comparing trimodulin with standard intravenous IgG (IVIG) preparation and investigating Fc receptor (FcR)-dependent functions by excluding IgM-mediated effects. Trimodulin demonstrated potent immunomodulatory, as well as anti-pathogenic effects in our neutrophil model (neutrophil-like HL-60 cells). The IgA component of trimodulin was shown to induce a strong FcαRI-dependent inhibitory immunoreceptor tyrosine-based activation motif (ITAMi) signaling, counteract lipopolysaccharide-induced inflammation and mediate phagocytosis of Staphylococcus aureus. The fine-tuned balance between immunomodulatory and anti-pathogenic effects of trimodulin were shown to be dose-dependent. Summarized, our data demonstrate the functional role of IgA in trimodulin, highlighting the importance of this immunoglobulin class in immunoglobulin therapy.
Collapse
Affiliation(s)
- Fabian Bohländer
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Sabrina Weißmüller
- Department of Translational Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Dennis Riehl
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Marcus Gutscher
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| | - Jörg Schüttrumpf
- Corporate R&D, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Stefanie Faust
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (F.B.); (D.R.); (M.G.)
| |
Collapse
|
67
|
Scavone M, Clerici B, Birocchi S, Mencarini T, Calogiuri M, Ghali C, Prati D, Bozzi S, Villa P, Cattaneo M, Podda GM. Platelet activation and modulation in thrombosis with thrombocytopenia syndrome associated with ChAdO×1 nCov-19 vaccine. Haematologica 2021; 106:3228-3231. [PMID: 34474550 PMCID: PMC8634168 DOI: 10.3324/haematol.2021.279345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Mariangela Scavone
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Bianca Clerici
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Simone Birocchi
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Tatiana Mencarini
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano
| | - Mariagrazia Calogiuri
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Claudia Ghali
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Daniele Prati
- Dipartimento di Medicina Trasfusionale e di Ematologia, IRCCS Fondazione Ca' Granda, Ospedale Maggiore Policlinico; Milano
| | - Silvia Bozzi
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano
| | - Paolo Villa
- U.O Medicina d'urgenza Sacco - Medico dell'ASST Fatebenefratelli Sacco, Milano
| | - Marco Cattaneo
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan
| | - Gian Marco Podda
- Divisione di Medicina Generale II, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan.
| |
Collapse
|
68
|
Kase Y, Takahashi H, Ito H, Kamata A, Amagai M, Yamagami J. Intravenous Ig Regulates Anti-Desmoglein 3 IgG Production in B220 - Antibody-Producing Cells in Mice with Pemphigus Vulgaris. J Invest Dermatol 2021; 142:1786-1792.e3. [PMID: 34848195 DOI: 10.1016/j.jid.2021.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/21/2021] [Accepted: 11/10/2021] [Indexed: 01/11/2023]
Abstract
Intravenous Ig (IVIG) is a treatment option for intractable cases of pemphigus vulgaris (PV), an autoimmune blistering disease caused by autoantibodies against desmoglein 3 (DSG3). To investigate the efficacy of IVIG on autoantibody secretion, we produced PV model mice by adoptive transfer of immunized Dsg3-/- splenocytes to Rag2-/- mice. We found that circulating anti-DSG3 IgG ELISA titer decreased in PV model mice after 5 days of treatment with IVIG compared with PBS-treated mice, whereas the F(ab')2 fragment did not suppress the anti-DSG3 IgG titer. enzyme-linked immunospot assay revealed that IVIG treatment reduced the frequency of anti-DSG3 antibody-secreting cells in the spleen but not in lymph nodes and bone marrow. Moreover, this reduction was observed only in the splenic B220- fraction but not in the B220+ fraction. Furthermore, IVIG decreased the serum levels of anti-DSG3 IgG, even after a significant reduction of its titer, owing to antibody-mediated CD20+ B cell depletion. In addition, IVIG suppressed anti-DSG3 IgG production in B220-CD138+ plasma cells derived from PV model mice ex vivo. These results indicate that IVIG reduced autoantibody production in B220- cells containing plasma cells in PV model mice, and this function may indicate one of the mechanisms of action of IVIG on PV.
Collapse
Affiliation(s)
- Yuko Kase
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Research and Development Division, Japan Blood Products Organization, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hiromi Ito
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Aki Kamata
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yamagami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
69
|
De Giacomo A, Gargano CD, Simone M, Petruzzelli MG, Pedaci C, Giambersio D, Margari L, Ruggieri M. B and T Immunoregulation: A New Insight of B Regulatory Lymphocytes in Autism Spectrum Disorder. Front Neurosci 2021; 15:732611. [PMID: 34776843 PMCID: PMC8581677 DOI: 10.3389/fnins.2021.732611] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction: Autism Spectrum Disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by a complex pathogenesis, by impairment social communication and interaction, and may also manifest repetitive patterns of behavior. Many studies have recognized an alteration of the immune response as a major etiological component in ASDs. Despite this, it is still unclear the variation of the function of the immune response. Aim: Our aim is to investigate the levels of immunological markers in peripheral blood of children with ASD such as: regulatory B and T cells, memory B and natural killer (NK) cells. Materials and Methods: We assessed various subsets of immune cells in peripheral blood (regulatory B and T cells, B-cell memory and natural killer cells) by multi-parametric flow cytometric analysis in 26 ASD children compared to 16 healthy controls (HCs) who matched age and gender. Results: No significant difference was observed between B-cell memory and NK cells in ASDs and HCs. Instead, regulatory B cells and T cells were decreased (p < 0.05) in ASD subjects when compared to HCs. Discussion: Regulatory B and T cells have a strategic role in maintaining the immune homeostasis. Their functions have been associated with the development of multiple pathologies especially in autoimmune diseases. According to our study, the immunological imbalance of regulatory B and T cells may play a pivotal role in the evolution of the disease, as immune deficiencies could be related to the severity of the ongoing disorder.
Collapse
Affiliation(s)
- Andrea De Giacomo
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Concetta Domenica Gargano
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Marta Simone
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Maria Giuseppina Petruzzelli
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Chiara Pedaci
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Donatella Giambersio
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Lucia Margari
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Maddalena Ruggieri
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
70
|
Kdimati S, Mullins CS, Linnebacher M. Cancer-Cell-Derived IgG and Its Potential Role in Tumor Development. Int J Mol Sci 2021; 22:ijms222111597. [PMID: 34769026 PMCID: PMC8583861 DOI: 10.3390/ijms222111597] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 12/18/2022] Open
Abstract
Human immunoglobulin G (IgG) is the primary component of the human serum antibody fraction, representing about 75% of the immunoglobulins and 10-20% of the total circulating plasma proteins. Generally, IgG sequences are highly conserved, yet the four subclasses, IgG1, IgG2, IgG3, and IgG4, differ in their physiological effector functions by binding to different IgG-Fc receptors (FcγR). Thus, despite a similarity of about 90% on the amino acid level, each subclass possesses a unique manner of antigen binding and immune complex formation. Triggering FcγR-expressing cells results in a wide range of responses, including phagocytosis, antibody-dependent cell-mediated cytotoxicity, and complement activation. Textbook knowledge implies that only B lymphocytes are capable of producing antibodies, which recognize specific antigenic structures derived from pathogens and infected endogenous or tumorigenic cells. Here, we review recent discoveries, including our own observations, about misplaced IgG expression in tumor cells. Various studies described the presence of IgG in tumor cells using immunohistology and established correlations between high antibody levels and promotion of cancer cell proliferation, invasion, and poor clinical prognosis for the respective tumor patients. Furthermore, blocking tumor-cell-derived IgG inhibited tumor cells. Tumor-cell-derived IgG might impede antigen-dependent cellular cytotoxicity by binding antigens while, at the same time, lacking the capacity for complement activation. These findings recommend tumor-cell-derived IgG as a potential therapeutic target. The observed uniqueness of Ig heavy chains expressed by tumor cells, using PCR with V(D)J rearrangement specific primers, suggests that this specific part of IgG may additionally play a role as a potential tumor marker and, thus, also qualify for the neoantigen category.
Collapse
|
71
|
Ganigara M, Sharma C, Bayry J. Unraveling the mechanisms of IVIG immunotherapy in MIS-C. CELL REPORTS MEDICINE 2021; 2:100431. [PMID: 34608458 PMCID: PMC8481087 DOI: 10.1016/j.xcrm.2021.100431] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In The Journal of Clinical Investigation, Zhu et al.1 report that intravenous immunoglobulin (IVIG) targets IL-1β+ neutrophils to exert anti-inflammatory effects in multisystem inflammatory syndrome in children (MIS-C), a post-infectious inflammatory condition associated with COVID-19.
Collapse
Affiliation(s)
- Madhusudan Ganigara
- Division of Pediatric Cardiology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Chetan Sharma
- Division of Pediatric Cardiology, Children's Hospital of San Antonio/Baylor College of Medicine, San Antonio, TX 78207, USA
| | - Jagadeesh Bayry
- Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, Kerala, India
| |
Collapse
|
72
|
Porritt RA, Binek A, Paschold L, Rivas MN, McArdle A, Yonker LM, Alter G, Chandnani HK, Lopez M, Fasano A, Van Eyk JE, Binder M, Arditi M. The autoimmune signature of hyperinflammatory multisystem inflammatory syndrome in children. J Clin Invest 2021; 131:e151520. [PMID: 34437303 PMCID: PMC8516454 DOI: 10.1172/jci151520] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) manifests as a severe and uncontrolled inflammatory response with multiorgan involvement, occurring weeks after SARS-CoV-2 infection. Here, we utilized proteomics, RNA sequencing, autoantibody arrays, and B cell receptor (BCR) repertoire analysis to characterize MIS-C immunopathogenesis and identify factors contributing to severe manifestations and intensive care unit admission. Inflammation markers, humoral immune responses, neutrophil activation, and complement and coagulation pathways were highly enriched in MIS-C patient serum, with a more hyperinflammatory profile in severe than in mild MIS-C cases. We identified a strong autoimmune signature in MIS-C, with autoantibodies targeted to both ubiquitously expressed and tissue-specific antigens, suggesting autoantigen release and excessive antigenic drive may result from systemic tissue damage. We further identified a cluster of patients with enhanced neutrophil responses as well as high anti-Spike IgG and autoantibody titers. BCR sequencing of these patients identified a strong imprint of antigenic drive with substantial BCR sequence connectivity and usage of autoimmunity-associated immunoglobulin heavy chain variable region (IGHV) genes. This cluster was linked to a TRBV11-2 expanded T cell receptor (TCR) repertoire, consistent with previous studies indicating a superantigen-driven pathogenic process. Overall, we identify a combination of pathogenic pathways that culminate in MIS-C and may inform treatment.
Collapse
Affiliation(s)
- Rebecca A. Porritt
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
| | - Angela McArdle
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lael M. Yonker
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Galit Alter
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MIT, MGH and Harvard, Cambridge, Massachusetts, USA
| | | | - Merrick Lopez
- Department of Pediatrics, Loma Linda University Hospital, California, USA
| | - Alessio Fasano
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
73
|
Kolahchi Z, Sohrabi H, Ekrami Nasab S, Jelodari Mamaghani H, Keyfari Alamdari M, Rezaei N. Potential therapeutic approach of intravenous immunoglobulin against COVID-19. Allergy Asthma Clin Immunol 2021; 17:105. [PMID: 34627384 PMCID: PMC8501925 DOI: 10.1186/s13223-021-00609-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
Since the outbreak of the novel coronavirus disease (COVID-19), the therapeutic and management options to reduce the burden of the COVID-19 disease are under investigation. IVIG therapy is used as an effective treatment for immunodeficient patients and patients with inflammatory or autoimmune conditions. The therapeutic effect of IVIG in COVID-19 patients has been investigated. But, the results are controversial and some studies reported no benefit of IVIG therapy. More clinical trials on the effect of IVIG therapy in COVID-19 patients should be performed to establish a certain conclusion about IVIG effectiveness.
Collapse
Affiliation(s)
- Zahra Kolahchi
- Students' International Committee of Medical Schools (SICoMS), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanye Sohrabi
- Students' International Committee of Medical Schools (SICoMS), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Ekrami Nasab
- Students' International Committee of Medical Schools (SICoMS), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hesan Jelodari Mamaghani
- Students' International Committee of Medical Schools (SICoMS), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Keyfari Alamdari
- Students' International Committee of Medical Schools (SICoMS), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
74
|
Esmaeilzadeh A, Rostami S, Yeganeh PM, Tahmasebi S, Ahmadi M. Recent advances in antibody-based immunotherapy strategies for COVID-19. J Cell Biochem 2021; 122:1389-1412. [PMID: 34160093 PMCID: PMC8427040 DOI: 10.1002/jcb.30017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 01/09/2023]
Abstract
The emergence of a new acute respiratory syndrome Coronavirus 2 (SARS-CoV-2), the cause of the 2019-nCOV disease (COVID-19), has caused a pandemic and a global health crisis. Rapid human-to-human transmission, even from asymptomatic individuals, has led to the quick spread of the virus worldwide, causing a wide range of clinical manifestations from cold-like symptoms to severe pneumonia, acute respiratory distress syndrome (ARDS), multiorgan injury, and even death. Therefore, using rapid and accurate diagnostic methods to identify the virus and subsequently select appropriate and effective treatments can help improvement of patients and control the pandemic. So far, various treatment regimens along with prophylactic vaccines have been developed to manage COVID-19-infected patients. Among these, antibody-based therapies, including neutralizing antibodies (against different parts of the virus), polyclonal and monoclonal antibodies, plasma therapy, and high-dose intravenous immunoglobulin (IVIG) have shown promising outcomes in accelerating and improving the treatment process of patients, avoiding the viral spreading widely, and managing the pandemic. In the current review paper, different types and applications of therapeutic antibodies in the COVID-19 treatment are comprehensively discussed.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, School of MedicineZanjan University of Medical SciencesZanjanIran
- Immunotherapy Research and Technology GroupZanjan University of Medical SciencesZanjanIran
| | - Samaneh Rostami
- Department of immunology, School of medicineZanjan University of Medical SciencesZanjanIran
| | - Pegah M. Yeganeh
- Department of immunology, School of medicineZanjan University of Medical SciencesZanjanIran
| | - Safa Tahmasebi
- Department of Immunology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Majid Ahmadi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
75
|
Stoian A, Bajko Z, Maier S, Cioflinc RA, Grigorescu BL, Moțățăianu A, Bărcuțean L, Balașa R, Stoian M. High-dose intravenous immunoglobulins as a therapeutic option in critical illness polyneuropathy accompanying SARS-CoV-2 infection: A case-based review of the literature (Review). Exp Ther Med 2021; 22:1182. [PMID: 34475972 PMCID: PMC8406741 DOI: 10.3892/etm.2021.10616] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/27/2021] [Indexed: 01/08/2023] Open
Abstract
The still ongoing COVID-19 pandemic has exposed the medical community to a number of major challenges. A significant number of patients require admission to intensive care unit (ICU) services due to severe respiratory, thrombotic and septic complications and require long-term hospitalization. Neuromuscular weakness is a common complication in critically ill patients who are treated in ICUs and are mechanically ventilated. This complication is frequently caused by critical illness myopathy (CIM) or critical illness polyneuropathy (CIP) and leads to difficulty in weaning from the ventilator. It is thought to represent an important neurologic manifestation of the systemic inflammatory response syndrome (SIRS). COVID-19 infection is known to trigger strong immune dysregulation, with an intense cytokine storm, as a result, the frequency of CIP is expected to be higher in this setting. The mainstay in the diagnosis of this entity beside the high level of clinical awareness is the electrophysiological examination that provides evidence of axonal motor and sensory polyneuropathy. The present article presents the case of a 54-year-old woman with severe COVID 19 infection who developed neuromuscular weakness, which turned out to be secondary to CIP and was treated successfully with a high dose of human intravenous immunoglobulins. Related to this case, we reviewed the relevant literature data regarding the epidemiology, pathophysiology and clinical features of this important complication and discussed also the treatment options and prognosis.
Collapse
Affiliation(s)
- Adina Stoian
- Department of Pathophysiology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Zoltan Bajko
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Smaranda Maier
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | | | - Bianca Liana Grigorescu
- Department of Pathophysiology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Anca Moțățăianu
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Laura Bărcuțean
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Rodica Balașa
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| | - Mircea Stoian
- Department of Anesthesiology and Intensive Therapy, ‘George Emil Palade’ University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania
| |
Collapse
|
76
|
de Cevins C, Luka M, Smith N, Meynier S, Magérus A, Carbone F, García-Paredes V, Barnabei L, Batignes M, Boullé A, Stolzenberg MC, Pérot BP, Charbit B, Fali T, Pirabakaran V, Sorin B, Riller Q, Abdessalem G, Beretta M, Grzelak L, Goncalves P, Di Santo JP, Mouquet H, Schwartz O, Zarhrate M, Parisot M, Bole-Feysot C, Masson C, Cagnard N, Corneau A, Brunaud C, Zhang SY, Casanova JL, Bader-Meunier B, Haroche J, Melki I, Lorrot M, Oualha M, Moulin F, Bonnet D, Belhadjer Z, Leruez M, Allali S, Gras-Leguen C, de Pontual L, Fischer A, Duffy D, Rieux-Laucat F, Toubiana J, Ménager MM. A monocyte/dendritic cell molecular signature of SARS-CoV-2-related multisystem inflammatory syndrome in children with severe myocarditis. MED 2021; 2:1072-1092.e7. [PMID: 34414385 PMCID: PMC8363470 DOI: 10.1016/j.medj.2021.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/12/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children is generally milder than in adults, but a proportion of cases result in hyperinflammatory conditions often including myocarditis. Methods To better understand these cases, we applied a multiparametric approach to the study of blood cells of 56 children hospitalized with suspicion of SARS-CoV-2 infection. Plasma cytokine and chemokine levels and blood cellular composition were measured, alongside gene expression at the bulk and single-cell levels. Findings The most severe forms of multisystem inflammatory syndrome in children (MIS-C) related to SARS-CoV-2 that resulted in myocarditis were characterized by elevated levels of pro-angiogenesis cytokines and several chemokines. Single-cell transcriptomics analyses identified a unique monocyte/dendritic cell gene signature that correlated with the occurrence of severe myocarditis characterized by sustained nuclear factor κB (NF-κB) activity and tumor necrosis factor alpha (TNF-α) signaling and associated with decreased gene expression of NF-κB inhibitors. We also found a weak response to type I and type II interferons, hyperinflammation, and response to oxidative stress related to increased HIF-1α and Vascular endothelial growth factor (VEGF) signaling. Conclusions These results provide potential for a better understanding of disease pathophysiology. Funding Agence National de la Recherche (Institut Hospitalo-Universitaire Imagine, grant ANR-10-IAHU-01; Recherche Hospitalo-Universitaire, grant ANR-18-RHUS-0010; Laboratoire d’Excellence ‘‘Milieu Intérieur,” grant ANR-10-LABX-69-01; ANR-flash Covid19 “AIROCovid” and “CoVarImm”), Institut National de la Santé et de la Recherche Médicale (INSERM), and the “URGENCE COVID-19” fundraising campaign of Institut Pasteur. Children with SARS-CoV-2 infection were initially thought to have only mild COVID-19 symptoms. However, several weeks into the first wave of SARS-CoV-2 infections, there was a surge of a postacute pathology called multisystem inflammatory syndrome in children (MIS-C). The authors recruited a cohort of children with suspicion of SARS-CoV-2 infection and uncovered hyperinflammation, hypoxic conditions, exacerbation of TNF-α signaling via NF-κB, and absence of responses to type I and type II IFN secretion in the most severe forms of MIS-C with severe myocarditis. This work led the authors to identify in monocytes and validate in peripheral blood mononuclear cells a molecular signature of 25 genes that allows discrimination of the most severe forms of MIS-C with myocarditis.
Collapse
Affiliation(s)
- Camille de Cevins
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Molecular Biology and Genomics, Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Marine Luka
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Nikaïa Smith
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
| | - Sonia Meynier
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Aude Magérus
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Francesco Carbone
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Víctor García-Paredes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Laura Barnabei
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Batignes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Alexandre Boullé
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Marie-Claude Stolzenberg
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Brieuc P Pérot
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Tinhinane Fali
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Vithura Pirabakaran
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Boris Sorin
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Quentin Riller
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Ghaith Abdessalem
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Beretta
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Ludivine Grzelak
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Pedro Goncalves
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - James P Di Santo
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Mohammed Zarhrate
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Christine Bole-Feysot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Cécile Masson
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Nicolas Cagnard
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Aurélien Corneau
- Sorbonne Université, UMS037, PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière CyPS, 75013 Paris, France
| | - Camille Brunaud
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Shen-Ying Zhang
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Brigitte Bader-Meunier
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Julien Haroche
- Department of Immunology and Infectious Disease (CIMI-Paris), Pitié-Salpêtrière University Hospital, Sorbonne Université, AP-HP, 75013 Paris, France
| | - Isabelle Melki
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Department of Pediatrics, Robert-Debré University Hospital, AP-HP, Université de Paris, Paris, France
| | - Mathie Lorrot
- Department of Pediatrics, Armand-Trousseau University Hospital, AP-HP, 75012 Paris, France
| | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Florence Moulin
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | | | | | - Marianne Leruez
- Virology Laboratory, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Slimane Allali
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
| | - Christèle Gras-Leguen
- Pediatric Department, Nantes University Hospital, CIC 1413, INSERM, 44000 Nantes, France
| | - Loïc de Pontual
- Department of Pediatrics, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Paris 13 University, Bondy, France
| | - Alain Fischer
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Université de Paris, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
- Collège de France, Paris, France
| | - Darragh Duffy
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Fredéric Rieux-Laucat
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Julie Toubiana
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
- Institut Pasteur, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
| | - Mickaël M Ménager
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| |
Collapse
|
77
|
Morotti F, Bracciolini G, Caorsi R, Cattaneo L, Gattorno M, Ravelli A, Felici E. Intravenous immunoglobulin for corticosteroid-resistant intestinal Henoch-Schönlein purpura: worth a controlled trial against corticosteroids? Rheumatology (Oxford) 2021; 60:3868-3871. [PMID: 34340243 DOI: 10.1093/rheumatology/keaa743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/01/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Henoch-Schönlein purpura (HScP) may present in children with severe, occasionally refractory, gastrointestinal (GI) involvement. The use of corticosteroids (CSs) is commonplace in the management of the disease, but to date no standardized protocol is available and, although rare, resistance to CS therapy may be challenging to clinicians. IVIG has been proposed as an effective alternative to CSs, but to date no controlled trial has been conducted to ascertain their real efficacy. We share our personal experience of successful IVIG treatment in two cases of GI HScP, comparing it with similar experiences reported in literature. METHODS Retrospective clinical data collection, comparison with available literature. RESULTS We describe two children with severe HScP GI vasculitis refractory to high-dose intravenous CSs that responded rapidly to IVIG administration, with complete recovery within a few days. Patient characteristics and response to IVIG administration were comparable to those of other previously reported cases. CONCLUSION Our observation confirms that IVIG may be useful in the treatment of CS-resistant HScP-related GI vasculitis in children, and highlights the need for more structured research, including a randomized trial against CSs, in order to ascertain their real effectiveness.
Collapse
Affiliation(s)
- Francesco Morotti
- SCDU of Pediatrics, Department of Health Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Giulia Bracciolini
- Pediatrics and Pediatric Emergency Unit, The Children Hospital, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Roberta Caorsi
- Clinica Pediatrica e Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Lorella Cattaneo
- Pediatrics and Pediatric Emergency Unit, The Children Hospital, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Marco Gattorno
- Clinica Pediatrica e Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Angelo Ravelli
- Clinica Pediatrica e Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Felici
- Pediatrics and Pediatric Emergency Unit, The Children Hospital, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.,'Umberto Bosio' Centre for Digestive Diseases, The Children Hospital, Azienda Ospedaliera Nazionale SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| |
Collapse
|
78
|
Abstract
ABSTRACT The ongoing coronavirus disease 2019 (COVID-19) pandemic has swept over the world and causes thousands of deaths. Although the clinical features of COVID-19 become much clearer than before, there are still further problems with the pathophysiological process and treatments of severe patients. One primary problem is with the paradoxical immune states in severe patients with COVID-19. Studies indicate that Severe Acute Respiratory Syndrome Coronavirus 2 can attack the immune system, manifested as a state of immunosuppression with a decrease in lymphocytes, whereas a state of hyperinflammation, presenting as elevated cytokine levels, is also detected in COVID-19. Therefore, discussing the specific status of immunity in COVID-19 will contribute to the understanding of its pathophysiology and the search for appropriate treatments. Here, we review all the available literature concerning the different immune states in COVID-19 and the underlying pathophysiological mechanisms. In addition, the association between immune states and the development and severity of disease as well as the impact on the selection of immunotherapy strategies are discussed in our review.
Collapse
Affiliation(s)
- Ye Liu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| |
Collapse
|
79
|
Liu L, Han C, Jiang M, Zhang T, Kang Q, Wang X, Wang P, Zhou F. Rapid and regenerable surface plasmon resonance determinations of biomarker concentration and biomolecular interaction based on tris-nitrilotriacetic acid chips. Anal Chim Acta 2021; 1170:338625. [PMID: 34090589 DOI: 10.1016/j.aca.2021.338625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
The tris-nitrilotriacetic acid (tris-NTA) chip has been used for surface plasmon resonance (SPR) kinetic studies involving histidine (His)-tagged proteins. However, its full potential, especially for analyte quantification in complex biological media, has not been realized due to a lack of systematic studies on the factors governing ligand immobilization, surface regeneration, and data analysis. We demonstrate that the tris-NTA chip not only retains His-tagged proteins more strongly than its mono-NTA counterpart, but also orients them more uniformly than protein molecules coupled to carboxymethylated dextran films. We accurately and rapidly quantified immunoglobulin (IgG) molecules in sera by using the initial association phase of their conjugation with His-tagged protein G densely immobilized onto the tris-NTA chip, and established criteria for selecting the optimal time for constructing the calibration curve. The method is highly reproducible (less than 2% RSD) and three orders of magnitude more sensitive than immunoturbidimetry. In addition, we found that the amount of His-protein immobilized is highly dependent on the protein isoelectric point (pI). Reliable kinetic data in a multi-channel SPR instrument can also be rapidly obtained by using a low density of immobilized His-tagged protein. The experimental parameters and procedures outlined in this study help expand the range of SPR applications involving His-tagged proteins.
Collapse
Affiliation(s)
- Luyao Liu
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Chaowei Han
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Meng Jiang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Tiantian Zhang
- University Hospital, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Qing Kang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China
| | - Xiaoying Wang
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology, Jinan, Shandong, 250353, PR China
| | - Pengcheng Wang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China.
| | - Feimeng Zhou
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, 250022, PR China.
| |
Collapse
|
80
|
Karnam A, Lacroix-Desmazes S, Kaveri SV, Bayry J. Vaccine-induced immune thrombotic thrombocytopenia: Consider IVIG batch in the treatment. J Thromb Haemost 2021; 19:1838-1839. [PMID: 33931935 PMCID: PMC8236969 DOI: 10.1111/jth.15361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 01/17/2023]
Affiliation(s)
- Anupama Karnam
- Centre de Recherché des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Sébastien Lacroix-Desmazes
- Centre de Recherché des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Srini V Kaveri
- Centre de Recherché des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Jagadeesh Bayry
- Centre de Recherché des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
- Indian Institute of Technology Palakkad, Palakkad, India
| |
Collapse
|
81
|
Naef B, Nilsson J, Wuethrich RP, Mueller TF, Schachtner T. Intravenous immunoglobulins do not prove beneficial to reduce alloimmunity among kidney transplant recipients with BKV-associated nephropathy. Transpl Int 2021; 34:1481-1493. [PMID: 33872427 DOI: 10.1111/tri.13882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/18/2021] [Accepted: 04/10/2021] [Indexed: 12/18/2022]
Abstract
Reduced immunosuppression during BKV-DNAemia has been associated with T-cell mediated rejection (TCMR), de novo donor-specific antibodies (DSA) and antibody-mediated rejection (ABMR). Intravenous immunoglobulins (IVIG) may reduce alloimmunity. We studied 860 kidney transplant recipients (KTRs) for the development of BKV-DNAuria and BKV-DNAemia (low-level <10 000 IE/ml, high-level >10 000 IE/ml). 52/131 KTRs with high-level BKV-DNAemia received IVIG. The HLA-related immunological risk was stratified by the Predicted Indirectly Recognizable HLA Epitopes (PIRCHE) algorithm. BKV-DNAuria only was observed in 86 KTRs (10.0%), low-level BKV-DNAemia in 180 KTRs (20.9%) and high-level BKV-DNAemia in 131 KTRs (15.2%). KTRs with low-level BKV-DNAemia showed significantly less TCMR compared to KTRs with high-level BKV-DNAemia (5.2% vs. 25.5%; P < 0.001) and no BKV-replication (13.2%; P = 0.014), lowest rates of de novo DSA (21.3%), ABMR (9.2%) and flattest glomerular filtration rate (GFR) slope (-0.8 ml/min). KTRs with low-level BKV-DNAemia showed significantly higher median (interquartile range) total PIRCHE if they developed TCMR [100.22 (72.6) vs. 69.52 (49.97); P = 0.020] or ABMR [128.86 (52.99) vs. 69.52 (49.96); P = 0.005]. Administration of IVIG did not shorten duration of BKV-DNAemia (P = 0.798) or reduce TCMR, de novo DSA and ABMR (P > 0.05). KTRs with low-level BKV-DNAemia showed best protection against alloimmunity, with a high number of PIRCHE co-determining the remaining risk. The administration of IVIG, however, was not beneficial in reducing alloimmunity.
Collapse
Affiliation(s)
- Bettina Naef
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Jakob Nilsson
- Division of Immunology, University Hospital Zurich, Zurich, Switzerland
| | | | - Thomas F Mueller
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Schachtner
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
82
|
Chun W, Kim Y, Park SH, Choi SJ. Thromboembolic complications following intravenous immunoglobulin therapy in immune-mediated neurological disorders. J Clin Neurosci 2021; 90:311-316. [PMID: 34275568 DOI: 10.1016/j.jocn.2021.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 05/24/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Minor adverse events of intravenous immunoglobulin (IVIg) include flu-like symptoms, eczematous skin reaction, electrolyte disturbance, and transient leukopenia. On rare occasions, serious complications such as aseptic meningitis, arrhythmia, decrease in blood pressure, and thromboembolic complications (TEC) have been described. The current study aimed to understand the frequency and clinical features of TEC related to IVIg administration in patients with immune-mediated neurological disorders. METHODS We conducted a retrospective chart review of hospitalized patients with immune-mediated neuromuscular or neuroimmunological disorders treated with IVIg from January 2018 to March 2020 in a single tertiary hospital. RESULTS During the study period, 61 patients were treated with a total of 364 IVIg infusions over 84 treatment courses. Among them, we identified 3 TEC cases that occurred during or after the completion of IVIg therapy: two patients with myasthenia gravis (F/60 and F/80) and one patient with Guillain-Barré syndrome (F/79) had undergone arterial TEC (two for ischemic stroke and one for pulmonary thromboembolism). The rates of TEC per patient, per treatment course, and per infusion were 4.91% (3/61), 3.57% (3/84), and 0.82% (3/364), respectively. CONCLUSION The risk of developing TEC upon receiving IVIg infusions is generally low in patients with immune-mediated neurological disorders; however, IVIg-related TEC should be cautiously monitored for in critically ill elderly patients with vascular risk factors, especially those suffering from myasthenic crisis.
Collapse
Affiliation(s)
- Woochang Chun
- Department of Neurology, Inha University Hospital, Incheon, Republic of Korea
| | - Yongchan Kim
- Department of Neurology, Inha University Hospital, Incheon, Republic of Korea
| | - Soo-Hyun Park
- Department of Neurology, Inha University Hospital, Incheon, Republic of Korea; Department of Critical Care Medicine, Inha University Hospital, Incheon, Republic of Korea
| | - Seok-Jin Choi
- Department of Neurology, Inha University Hospital, Incheon, Republic of Korea; Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
83
|
Bohländer F, Riehl D, Weißmüller S, Gutscher M, Schüttrumpf J, Faust S. Immunomodulation: Immunoglobulin Preparations Suppress Hyperinflammation in a COVID-19 Model via FcγRIIA and FcαRI. Front Immunol 2021; 12:700429. [PMID: 34177967 PMCID: PMC8223875 DOI: 10.3389/fimmu.2021.700429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
The rapid spread of SARS-CoV-2 has induced a global pandemic. Severe forms of COVID-19 are characterized by dysregulated immune response and "cytokine storm". The role of IgG and IgM antibodies in COVID-19 pathology is reasonably well studied, whereas IgA is neglected. To improve clinical outcome of patients, immune modulatory drugs appear to be beneficial. Such drugs include intravenous immunoglobulin preparations, which were successfully tested in severe COVID-19 patients. Here we established a versatile in vitro model to study inflammatory as well as anti-inflammatory processes by therapeutic human immunoglobulins. We dissect the inflammatory activation on neutrophil-like HL60 cells, using an immune complex consisting of latex beads coated with spike protein of SARS-CoV-2 and opsonized with specific immunoglobulins from convalescent plasma. Our data clarifies the role of Fc-receptor-dependent phagocytosis via IgA-FcαRI and IgG-FcγR for COVID-19 disease followed by cytokine release. We show that COVID-19 associated inflammation could be reduced by addition of human immunoglobulin preparations (IVIG and trimodulin), while trimodulin elicits stronger immune modulation by more powerful ITAMi signaling. Besides IgG, the IgA component of trimodulin in particular, is of functional relevance for immune modulation in this assay setup, highlighting the need to study IgA mediated immune response.
Collapse
Affiliation(s)
- Fabian Bohländer
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | - Dennis Riehl
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | - Sabrina Weißmüller
- Corporate R&D, Biotest AG, Dreieich, Germany
- Department of Translational Research, Preclinical Research, Corporate R&D, Biotest AG, Dreieich, Germany
| | - Marcus Gutscher
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| | | | - Stefanie Faust
- Department of Analytical Development and Validation, Corporate R&D, Biotest AG, Dreieich, Germany
- Corporate R&D, Biotest AG, Dreieich, Germany
| |
Collapse
|
84
|
Kobak S. The Perfect Storm: A Rheumatologist's Point of View on COVID-19 Infection. Curr Rheumatol Rev 2021; 17:141-152. [PMID: 33121412 DOI: 10.2174/1573397116666201029155105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/20/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022]
Abstract
The new coronavirus infection (Covid-19) is a pandemic that has affected the whole world and progresses with high morbidity and mortality. It has a high contagion rate and a course capable of rapid lung involvement with severe acute respiratory distress syndrome (ARDS) and pulmonary insufficiency. A severe clinical picture develops as a result of a "perfect cytokine storm" which results from possible immunological mechanisms triggered by the viral infection. Immune system dysregulation and possible autoinflammatory and autoimmune mechanisms are responsible for a higher amount of cytokines release from immune cells. Although no clear treatment of Covid-19 infection has emerged yet, it is argued that some disease-modifying anti-rheumatic drugs (DMARDs) may be effective in addition to anti-viral treatments. These drugs (anti-malarial drugs, colchicum dispert, biologics) have been well known to rheumatologists for years because they are used in the treatment of many inflammatory rheumatologic diseases. Another important issue is whether DMARDs, which can cause severe immunosuppression, pose a risk for Covid-19 infection and whether they have been discontinued beforehand. Although there are insufficient data on this subject, considering the risk of disease reactivation, patients may continue their DMARDs treatment under the supervision of a rheumatologist. In this article, the possible immunological mechanisms in the pathogenesis of Covid-19 infection and the efficacy and safety of various DMARDs used in the treatment are discussed from a rheumatologist's perspective in the light of recent literature data.
Collapse
Affiliation(s)
- Senol Kobak
- Department of Internal Medicine and Rheumatology, Faculty of Medicine, Istinye University, Liv Hospital, Istanbul, Turkey
| |
Collapse
|
85
|
Kim YS, Yang HJ, Kee SJ, Choi I, Ha K, Ki KK, Jeong IS, Cho HJ. The "Intermediate" CD14 + CD16 + monocyte subpopulation plays a role in IVIG responsiveness of children with Kawasaki disease. Pediatr Rheumatol Online J 2021; 19:76. [PMID: 34059085 PMCID: PMC8165978 DOI: 10.1186/s12969-021-00573-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Kawasaki disease (KD) is an acute, self-limited febrile illness of unknown cause. Intravenous immunoglobulin (IVIG)-resistance are related to greater risk for permanent cardiac complications. We aimed to determine the correlation between monocytes and the phenotype of KD in relation to IVIG responsiveness in children. MATERIALS AND METHODS The study cohort included 62 patients who were diagnosed with KD, 20 non febrile healthy controls (NFC), and 15 other febrile controls (OFC). In all enrolled patients, blood was taken at least 4 times and laboratory tests were performed. In addition, subtypes of monocytes were characterized via flow cytometry. RESULTS The numbers of intermediate monocytes were significantly lower in IVIG-resistant group compared to IVIG-responsive group before IVIG infusion (p < 0.0001). After infusion, intermediate monocytes decreased in the responsive group, while a trend of increase was observed in the resistant group. Only intermediate monocytes were significant in logistic regression with adjusted OR of 0.001 and p value of 0.03. CONCLUSIONS CD14 + CD16 + intermediate monocyte may play an important role in IVIG responsiveness among KD children. Low starting levels of intermediate monocytes, followed by a dramatic increase post-IVIG infusion during acute phase of KD are associated with IVIG-resistance. Functional studies on intermediate monocyte may help to reveal the pathophysiology.
Collapse
Affiliation(s)
- Yi Seul Kim
- grid.14005.300000 0001 0356 9399Department of Pediatric, Chonnam National University Children’s Hospital, 42 Jaebong ro, Gwangju, South Korea ,grid.255649.90000 0001 2171 7754Department of Pediatrics, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Hyun Jin Yang
- grid.14005.300000 0001 0356 9399Chonnam National University Medical School, 42 Jaebong ro, Gwangju, South Korea
| | - Seung-Jung Kee
- grid.14005.300000 0001 0356 9399Chonnam National University Medical School, 42 Jaebong ro, Gwangju, South Korea ,grid.411602.00000 0004 0647 9534Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Insu Choi
- grid.14005.300000 0001 0356 9399Department of Pediatric, Chonnam National University Children’s Hospital, 42 Jaebong ro, Gwangju, South Korea
| | - Kisoo Ha
- grid.411134.20000 0004 0474 0479Department of Pediatrics, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Katrina K Ki
- grid.415184.d0000 0004 0614 0266Critical Care Research Group,, The Prince Charles Hospital, Queensland Chermside, Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, The University of Queensland, St. Lucia, Queensland Australia
| | - In Seok Jeong
- Chonnam National University Medical School, 42 Jaebong ro, Gwangju, South Korea. .,Deparment of Cardiothoracic Surgery, Chonnam National University Hospital and Medical School, 42 Jaebong ro, Gwangju, South Korea.
| | - Hwa Jin Cho
- Department of Pediatric, Chonnam National University Children's Hospital, 42 Jaebong ro, Gwangju, South Korea. .,Chonnam National University Medical School, 42 Jaebong ro, Gwangju, South Korea.
| |
Collapse
|
86
|
Tran VL, Parsons S, Nuibe A. The Trilogy of SARS-CoV-2 in Pediatrics (Part 2): Multisystem Inflammatory Syndrome in Children. J Pediatr Pharmacol Ther 2021; 26:318-338. [PMID: 34035676 DOI: 10.5863/1551-6776-26.4.318] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022]
Abstract
Multisystem Inflammatory Syndrome in Children (MIS-C) was first recognized as a novel illness in 2020 with manifestations similar to other hyperinflammatory syndromes, such as Kawasaki disease or macrophage activation syndrome. Severity varies from a self-limited febrile illness to shock requiring inotropes and mechanical ventilation. Gastrointestinal symptoms and persistent fevers are the most common clinical symptoms, with the addition of cardiac manifestations inclusive of ventricular dysfunction and coronary artery aneurysms. With no controlled trials or comparative effectiveness studies evaluating treatment of MIS-C to date, current treatment with immunomodulatory agents has mainly been derived from previous experience treating Kawasaki disease. This article provides a comprehensive review summarizing published data for the evaluation and management of MIS-C, with a focus on pharmacotherapy treatment considerations.
Collapse
|
87
|
Immune-Based Therapy for COVID-19. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:449-468. [PMID: 33973194 DOI: 10.1007/978-3-030-63761-3_26] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel zoonotic virus identified as the cause of coronavirus disease 2019 (COVID-19) that has crossed species and infected humans. In order to develop new insights on the immune-based treatments against this disease, it is vital to understand the immunopathology of the COVID-19, implications of the immune response to SARS-CoV-2, and immune dysfunction in response to SARS-CoV-2. There is no approved drug for the treatment of COVID-19. It is, thus, promising to design immune-based treatments that inhibit the infectious mechanism of the virus, improve the inadequate immune response, or regulate the hyperactivated immune response in severely ill patients. According to the antiviral immune response against the virus, antibody-based immunotherapies of COVID-19 include injection of convalescent plasma from recovered patients, high-dose intravenous immunoglobulins (IVIG), monoclonal antibodies, and polyclonal antibodies. Also, cell-based treatment, vaccine-based approaches, cytokine-based immunotherapy, immune checkpoint inhibitors, JAK inhibitors, decoy receptors, and immunosuppressive drugs are discussed in this chapter.
Collapse
|
88
|
Boolean analysis of the transcriptomic data to identify novel biomarkers of IVIG response. Autoimmun Rev 2021; 20:102850. [PMID: 33971345 DOI: 10.1016/j.autrev.2021.102850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022]
Abstract
Intravenous immunoglobulin (IVIG) is used to treat several autoimmune and inflammatory diseases, but some patients are refractory to IVIG and require alternative treatments. Identifying a biomarker that could segregate IVIG responders from non-responders has been a subject of intense research. Unfortunately, previous transcriptomic studies aimed at addressing IVIG resistance have failed to predict a biomarker that could identify IVIG-non-responders. Therefore, we used a novel data mining technique on the publicly available transcriptomic data of Kawasaki disease (KD) patients treated with IVIG to identify potential biomarkers of IVIG response. By studying the boolean patterns hidden in the expression profiles of KD patients undergoing IVIG therapy, we have identified new metabolic pathways implicated in IVIG resistance in KD. These pathways could be used as biomarkers to segregate IVIG non-responders from responders prior to IVIG infusion. Also, boolean analysis of the transcriptomic data could be further extended to identify a universal biomarker that might predict IVIG response in other autoimmune diseases.
Collapse
|
89
|
Meridor K, Shoenfeld Y, Tayer-Shifman O, Levy Y. Lupus acute cardiomyopathy is highly responsive to intravenous immunoglobulin treatment: Case series and literature review. Medicine (Baltimore) 2021; 100:e25591. [PMID: 33950936 PMCID: PMC8104142 DOI: 10.1097/md.0000000000025591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Intravenous immunoglobulin (IVIg) is currently used with considerable success for the treatment of many autoimmune diseases, including systemic lupus erythematosus (SLE). Among its various indications, IVIg has also been found to be beneficial in myocarditis, whether or not it is associated with an autoimmune disease. Nevertheless, data regarding IVIg treatment for myocarditis/cardiomyopathy in patients with SLE are sparse. The objective of this case series was to describe our experience with IVIg as a treatment for lupus myocarditis and to review the literature for IVIg for this indication. PATIENT CONCERNS We report 5 female patients with SLE, who presented with signs of acute heart failure including pulmonary congestion and arrhythmias. DIAGNOSIS Echocardiography demonstrated new reduced left ventricular ejection fraction of 20% to 30%. Two patients underwent coronary artery angiography, which demonstrated normal coronary arteries, supporting the diagnosis of myocarditis or nonischemic cardiomyopathy. INTERVENTIONS High-dose IVIg treatment was initiated in all 5 patients. OUTCOMES Following the treatment, clinical and echocardiographic improvement in cardiac function occurred within a few days to 1 month. This dramatic improvement persisted for several years. CONCLUSION Based on our case series, we believe that IVIg has an important role in the management of lupus acute cardiomyopathy. This safe, well-tolerated optional treatment should be considered, especially in severe cases.
Collapse
Affiliation(s)
- Katya Meridor
- Department of Internal Medicine E, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel HaShomer
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oshrat Tayer-Shifman
- Rheumatology Unit, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yair Levy
- Department of Internal Medicine E, Meir Medical Center, Kfar Saba
- Affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
90
|
Fatima N, Kaushik V, Ayoub A. A Narrative Review of a Pulmonary Aerosolized Formulation or a Nasal Drop Using Sera Containing Neutralizing Antibodies Collected from COVID-19-Recovered Patients as a Probable Therapy for COVID-19. IRANIAN JOURNAL OF MEDICAL SCIENCES 2021; 46:151-168. [PMID: 34083848 PMCID: PMC8163704 DOI: 10.30476/ijms.2020.86417.1624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/29/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) emerged as a new contagion during December 2019, since which time it has triggered a rampant spike in fatality rates worldwide due to insufficient medical treatments and a lack of counteragents and prompted the World Health Organization to declare COVID-19 a public health emergency. It is, therefore, vital to accelerate the screening of new molecules or vaccines to win the battle against this pandemic. Experiences from previous epidemiological data on coronaviruses guide investigators in designing and exploring new compounds for a safe and cost-effective treatment. Several reports on the severe acute respiratory syndrome (SARS) epidemic indicate that severe acute respiratory syndrome coronavirus (SARS-CoV) and the novel COVID-19 use angiotensin-converting enzyme 2 (ACE2) as a receptor for binding to the host cell in the lung epithelia through the spike protein on their virion surface. ACE2 is a mono-carboxypeptidase best known for cleaving major peptides and substrates. Its degree in human airway epithelia positively correlates with coronavirus infection. The treatment approach can be the neutralization of the virus entering lung epithelial cells by using sera containing antibodies collected from COVID-19-recovered patients. Hence, we herein propose a pulmonary aerosolized formulation or a nasal drop using sera, which contain antibodies to prevent, treat, or immunize against COVID-19 infection.
Collapse
Affiliation(s)
- Nishat Fatima
- School of Pharmacy, Al-Hawash Private University, Homs, Syria
| | | | - Amjad Ayoub
- School of Pharmacy, Al-Hawash Private University, Homs, Syria
| |
Collapse
|
91
|
Peters K, Peters M. The Role of Lectin Receptors and Their Ligands in Controlling Allergic Inflammation. Front Immunol 2021; 12:635411. [PMID: 33995354 PMCID: PMC8119883 DOI: 10.3389/fimmu.2021.635411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/16/2021] [Indexed: 01/29/2023] Open
Abstract
More than fifty c-type lectin receptors (CLR) are known and have been identified so far. Moreover, we know the group of galectins and sialic acid-binding immunoglobulin-type lectins that also belong to the carbohydrate-binding receptors of the immune system. Thus, the lectin receptors form the largest receptor family among the pathogen recognition receptors. Similar to the toll-like receptors (TLRs), the CLR do not only recognize foreign but also endogenous molecules. In contrast to TLRs, which have a predominantly activating effect on the immune system, lectin receptors also mediate inhibitory signals. They play an important role in innate and adaptive immunity for the induction, regulation and shaping of the immune response. The hygiene hypothesis links enhanced infection to protection from allergic disease. Yet, the microbial substances that are responsible for mediating this allergy-protective activity still have to be identified. Microbes contain both ligands binding to TLRs and carbohydrates that are recognized by CLR and other lectin receptors. In the current literature, the CLR are often recognized as the ‘bad guys’ in allergic inflammation, because some glycoepitopes of allergens have been shown to bind to CLR, facilitating their uptake and presentation. On the other hand, there are many reports revealing that sugar moieties are involved in immune regulation. In this review, we will summarize what is known about the role of carbohydrate interaction with c-type lectins and other sugar-recognizing receptors in anti-inflammation, with a special focus on the regulation of the allergic immune response.
Collapse
Affiliation(s)
- Karin Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
92
|
Kim MY, Brennan DC. Therapies for Chronic Allograft Rejection. Front Pharmacol 2021; 12:651222. [PMID: 33935762 PMCID: PMC8082459 DOI: 10.3389/fphar.2021.651222] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Remarkable advances have been made in the pathophysiology, diagnosis, and treatment of antibody-mediated rejection (ABMR) over the past decades, leading to improved graft outcomes. However, long-term failure is still high and effective treatment for chronic ABMR, an important cause of graft failure, has not yet been identified. Chronic ABMR has a relatively different phenotype from active ABMR and is a slowly progressive disease in which graft injury is mainly caused by de novo donor specific antibodies (DSA). Since most trials of current immunosuppressive therapies for rejection have focused on active ABMR, treatment strategies based on those data might be less effective in chronic ABMR. A better understanding of chronic ABMR may serve as a bridge in establishing treatment strategies to improve graft outcomes. In this in-depth review, we focus on the pathophysiology and characteristics of chronic ABMR along with the newly revised Banff criteria in 2017. In addition, in terms of chronic ABMR, we identify the reasons for the resistance of current immunosuppressive therapies and look at ongoing research that could play a role in setting better treatment strategies in the future. Finally, we review non-invasive biomarkers as tools to monitor for rejection.
Collapse
Affiliation(s)
| | - Daniel C. Brennan
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
93
|
Lalmahomed TA, Walter IJ, Lely AT, Bloemenkamp KWM, Kooiman J, Limper M. On the use of intravenous immunoglobulins for the treatment of the antiphospholipid syndrome - A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102828. [PMID: 33866065 DOI: 10.1016/j.autrev.2021.102828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/03/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Tariq Anwar Lalmahomed
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Isabel Johanna Walter
- Department of Gynaecology and Obstetrics, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne Titia Lely
- Department of Gynaecology and Obstetrics, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Judith Kooiman
- Department of Gynaecology and Obstetrics, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maarten Limper
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
94
|
Kaplan AC. Noteworthy Neurological Manifestations Associated With COVID-19 Infection. Cureus 2021; 13:e14391. [PMID: 33987051 PMCID: PMC8110288 DOI: 10.7759/cureus.14391] [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] [Indexed: 11/22/2022] Open
Abstract
March 11, 2020 marked the start of the coronavirus disease 2019 (COVID-19) pandemic. COVID-19, caused by severe acute respiratory syndrome coronavirus 2, was being reported as a severe respiratory illness. However, since the recognition of this novel virus, there has been a constant realization that it may present or manifest in a multitude of ways. At first, the typical signs and symptoms were what one would expect from a respiratory virus: cough, shortness of breath, and fever. However, as the disease became more prevalent, neurologic symptoms were reported such as headaches, hypogeusia, and hyposmia. This case report aims to add to the growing body of neurologic manifestations by presenting two cases, Bell’s palsy and Guillain-Barre syndrome. Each case involves flaccid paralysis as the primary presentation.
Collapse
Affiliation(s)
- Adam C Kaplan
- Internal Medicine, St. Francis Medical Center, Trenton, USA
| |
Collapse
|
95
|
Varadé J, Magadán S, González-Fernández Á. Human immunology and immunotherapy: main achievements and challenges. Cell Mol Immunol 2021; 18:805-828. [PMID: 32879472 PMCID: PMC7463107 DOI: 10.1038/s41423-020-00530-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
The immune system is a fascinating world of cells, soluble factors, interacting cells, and tissues, all of which are interconnected. The highly complex nature of the immune system makes it difficult to view it as a whole, but researchers are now trying to put all the pieces of the puzzle together to obtain a more complete picture. The development of new specialized equipment and immunological techniques, genetic approaches, animal models, and a long list of monoclonal antibodies, among many other factors, are improving our knowledge of this sophisticated system. The different types of cell subsets, soluble factors, membrane molecules, and cell functionalities are some aspects that we are starting to understand, together with their roles in health, aging, and illness. This knowledge is filling many of the gaps, and in some cases, it has led to changes in our previous assumptions; e.g., adaptive immune cells were previously thought to be unique memory cells until trained innate immunity was observed, and several innate immune cells with features similar to those of cytokine-secreting T cells have been discovered. Moreover, we have improved our knowledge not only regarding immune-mediated illnesses and how the immune system works and interacts with other systems and components (such as the microbiome) but also in terms of ways to manipulate this system through immunotherapy. The development of different types of immunotherapies, including vaccines (prophylactic and therapeutic), and the use of pathogens, monoclonal antibodies, recombinant proteins, cytokines, and cellular immunotherapies, are changing the way in which we approach many diseases, especially cancer.
Collapse
Affiliation(s)
- Jezabel Varadé
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Susana Magadán
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - África González-Fernández
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain.
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
96
|
Hart SJ, Worley G, Kishnani PS, Van Mater H. Case Report: Improvement Following Immunotherapy in an Individual With Seronegative Down Syndrome Disintegrative Disorder. Front Neurol 2021; 12:621637. [PMID: 33841297 PMCID: PMC8032932 DOI: 10.3389/fneur.2021.621637] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Down syndrome disintegrative disorder (DSDD) is a condition of unknown etiology characterized by acute cognitive decline, catatonia, insomnia, and autistic features in individuals with Down syndrome. A prior report of four patients with DSDD suggested a potential autoimmune etiology based on the presence of autoantibodies and on successful treatment with immunotherapy that included intravenous immunoglobulin (IVIG). Herein, we present the case of an 8-year old girl who developed acute cognitive decline to a dementia-like state, insomnia, catatonia, and autistic features. In contrast to the four patients with DSDD above, she had no evidence of autoimmunity and presented at a younger age. Given the gravity of her acute deterioration and the exclusion of other etiologies, she was treated with immunotherapy presumptively. She responded with near complete resolution of symptoms, but demonstrated a pattern of mild decline as she approached each monthly dosing of IVIG and steroids, reversed by treatment. Mycophenolate mofetil (MMF) was therefore added, with stability throughout the month and the ability to taper off IVIG. After stopping IVIG, she had a mild recurrence of symptoms that again resolved with repeat IVIG followed by tapering off. Outcome was assessed at 2.5 years after presentation, at which time she was back to her premorbid condition, except for persistent tics off immunotherapy. This case supports the contention that patients with a rapid onset of severe symptoms consistent with DSDD, who have a thorough evaluation with the exclusion of other etiologies, may warrant a trial of immunotherapy with steroids, IVIG and/or other agents like MMF even in the absence of evidence of autoimmunity on standard evaluation.
Collapse
Affiliation(s)
- Sarah J Hart
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, United States
| | - Gordon Worley
- Department of Pediatrics, Division of Pediatric Neurology, Duke University Medical Center, Durham, NC, United States
| | - Priya S Kishnani
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, United States
| | - Heather Van Mater
- Department of Pediatrics, Division of Pediatric Rheumatology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
97
|
Cui M, Huang J, Zhang S, Liu Q, Liao Q, Qiu X. Immunoglobulin Expression in Cancer Cells and Its Critical Roles in Tumorigenesis. Front Immunol 2021; 12:613530. [PMID: 33841396 PMCID: PMC8024581 DOI: 10.3389/fimmu.2021.613530] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
Traditionally, immunoglobulin (Ig) was believed to be produced by only B-lineage cells. However, increasing evidence has revealed a high level of Ig expression in cancer cells, and this Ig is named cancer-derived Ig. Further studies have shown that cancer-derived Ig shares identical basic structures with B cell-derived Ig but exhibits several distinct characteristics, including restricted variable region sequences and aberrant glycosylation. In contrast to B cell-derived Ig, which functions as an antibody in the humoral immune response, cancer-derived Ig exerts profound protumorigenic effects via multiple mechanisms, including promoting the malignant behaviors of cancer cells, mediating tumor immune escape, inducing inflammation, and activating the aggregation of platelets. Importantly, cancer-derived Ig shows promising potential for application as a diagnostic and therapeutic target in cancer patients. In this review, we summarize progress in the research area of cancer-derived Ig and discuss the perspectives of applying this novel target for the management of cancer patients.
Collapse
Affiliation(s)
- Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shenghua Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
98
|
Intravenous Immunoglobulin for Treatment of Patients with COVID-19: A Case-control Study. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2021. [DOI: 10.5812/archcid.108068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: It seems that the risk of developing complications associated with coronavirus disease 2019 (COVID-19) is higher among individuals with weakened immune systems. Objectives: Therefore, this study was carried out to determine the effectiveness of intravenous immunoglobulin (IVIG) for the treatment of patients not entering the intubation phase compared to those entering the intubation phase. Methods: This descriptive case-control study was performed on 26 patients with COVID-19 referring to Imam Reza hospital in Mashhad, Iran, in March 2020. For subjects with COVID-19 not responding to the standard three-drug protocol (i.e., ribavirin, hydroxychloroquine, and lopinavir/ritonavir), three doses of IVIG (0.4 g/kg/day) were added to the protocol. The patients were divided into two groups of subjects not entering the intubation phase and those entering the intubation phase and compared in terms of different variables. Results: The comparison of laboratory findings showed a significant difference before and after receiving IVIG regarding oxygen saturation (P < 0.005), white blood cell (P = 0.001), hemoglobin level (P = 0.0002), lymphocyte count (P = 0.03), and C-reactive protein (P = 0.001). In general, 53.8% and 46.2% of the patients were discharged and expired, respectively. All the subjects not entering the intubation phase were recovered; nevertheless, only one case entering the intubation phase was recovered, and 92.3% of the patients expired. A significant difference was observed between the patients not entering the intubation phase and those entering the intubation phase in terms of mortality (χ2 = 22.28; P < 0.005). Conclusions: In summary, the obtained results of the current study confirmed the therapeutic effects of IVIG on patients with COVID-19. Moreover, better treatment results, shorter hospital stay, and lower mortality rates were observed among COVID-19 patients who did not enter the intubation phase in comparison with those entering the intubation phase.
Collapse
|
99
|
Bartoli A, Gabrielli F, Alicandro T, Nascimbeni F, Andreone P. COVID-19 treatment options: a difficult journey between failed attempts and experimental drugs. Intern Emerg Med 2021; 16:281-308. [PMID: 33398609 PMCID: PMC7781413 DOI: 10.1007/s11739-020-02569-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 11/10/2020] [Indexed: 01/31/2023]
Abstract
Since its outbreak in China in December 2019 a novel Coronavirus, named SARS-CoV-2, has spread worldwide causing many cases of severe pneumonia, referred to as COVID-19 disease, leading the World Health Organization to declare a pandemic emergency in March 2020. Up to now, no specific therapy against COVID-19 disease exists. This paper aims to review COVID-19 treatment options currently under investigation. We divided the studied drugs into three categories (antiviral, immunomodulatory and other drugs). For each molecule, we discussed the putative mechanisms by which the drug may act against SARS-CoV-2 or may affect COVID-19 pathogenesis and the main clinical studies performed so far. The published clinical studies suffer from methodological limitations due to the emergency setting in which they have been conducted. Nevertheless, it seems that the timing of administration of the diverse categories of drugs is crucial in determining clinical efficacy. Antiviral drugs, in particular Remdesivir, should be administered soon after symptoms onset, in the viraemic phase of the disease; whereas, immunomodulatory agents, such as tocilizumab, anakinra and steroids, may have better results if administered in pneumonia/hyperinflammatory phases. Low-molecular-weight heparin may also have a role when facing COVID-19-related coagulopathy. Up to now, treatment choices have been inferred from the experience with other coronaviruses or viral infection outbreaks. Hopefully, in the near future, new treatment strategies will be available thanks to increased knowledge on SARS-CoV2 virus and COVID-19 pathogenesis. In the meanwhile, further well-designed clinical trials are urgently needed to establish a standard of care in COVID-19 disease.
Collapse
Affiliation(s)
- Alessandra Bartoli
- Division of Internal Medicine and Metabolism, Department of Internal Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | - Filippo Gabrielli
- Division of Internal Medicine and Metabolism, Department of Internal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Tatiana Alicandro
- Division of Internal Medicine and Metabolism, Department of Internal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Nascimbeni
- Division of Internal Medicine and Metabolism, Department of Internal Medicine, University Hospital of Modena, Modena, Italy
| | - Pietro Andreone
- Division of Internal Medicine and Metabolism, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
100
|
Bhardwaj A, Sapra L, Saini C, Azam Z, Mishra PK, Verma B, Mishra GC, Srivastava RK. COVID-19: Immunology, Immunopathogenesis and Potential Therapies. Int Rev Immunol 2021; 41:171-206. [PMID: 33641587 PMCID: PMC7919479 DOI: 10.1080/08830185.2021.1883600] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/09/2020] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
The Coronavirus Disease-2019 (COVID-19) imposed public health emergency and affected millions of people around the globe. As of January 2021, 100 million confirmed cases of COVID-19 along with more than 2 million deaths were reported worldwide. SARS-CoV-2 infection causes excessive production of pro-inflammatory cytokines thereby leading to the development of "Cytokine Storm Syndrome." This condition results in uncontrollable inflammation that further imposes multiple-organ-failure eventually leading to death. SARS-CoV-2 induces unrestrained innate immune response and impairs adaptive immune responses thereby causing tissue damage. Thus, understanding the foremost features and evolution of innate and adaptive immunity to SARS-CoV-2 is crucial in anticipating COVID-19 outcomes and in developing effective strategies to control the viral spread. In the present review, we exhaustively discuss the sequential key immunological events that occur during SARS-CoV-2 infection and are involved in the immunopathogenesis of COVID-19. In addition to this, we also highlight various therapeutic options already in use such as immunosuppressive drugs, plasma therapy and intravenous immunoglobulins along with various novel potent therapeutic options that should be considered in managing COVID-19 infection such as traditional medicines and probiotics.
Collapse
Affiliation(s)
- Asha Bhardwaj
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Leena Sapra
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Chaman Saini
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Zaffar Azam
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Pradyumna K. Mishra
- Department of Molecular Biology, ICMR-NIREH, Nehru Hospital Building, Gandhi Medical College Campus, Bhopal, India
| | - Bhupendra Verma
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Gyan C. Mishra
- Lab # 1, National Centre for Cell Science (NCCS), Savitribai Phule Pune University Campus, Pune, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|