Pub Date : 2022-06-01Epub Date: 2021-10-16DOI: 10.1080/25785826.2021.1963190
Shizhu Jin, Narayan Nepal, Yang Gao
Helicobacter pylori (HP) is the primary etiologic factor that induces events in the immune system that lead to peptic ulcers. Toll-like receptors (TLRs) are an important part of the innate immune system, as they play pivotal roles in pathogen-associated molecular pattern (PAMP) recognition of HP as well host-associated damage-associated molecular patterns (DAMPs). Recent advancements such as COX-2 production, LPS recognition through TLR2, CagL, and CagY protein of HP activating TLR5, TLR9 activation via type IV secretion system (T4SS) using DNA transfer, TLR polymorphisms, their adaptor molecules, cytokines, and other factors play a significant role in PUD. Thus, some novel PUD treatments including Chuyou Yuyang granules, function by TLR4/NF-κB signaling pathway suppression and TNF-α and IL-18 inhibition also rely on TLR signaling. Similarly glycyrrhetinic acid (GA) treatment activates TLR-4 in Ana-1 cells not via TRIF, but via MYD88 expression, which is significantly upregulated to cure PUD. Therefore, understanding TLR signaling complexity and its resultant immune modulation after host-pathogen interactions is pivotal to drug and vaccine development for other diseases as well including cancer and recent pandemic COVID-19. In this review, we summarize the TLRs and HP interaction; its pathophysiology-related signaling pathways, polymorphisms, and pharmaceutical approaches toward PUD.
{"title":"The role of toll-like receptors in peptic ulcer disease.","authors":"Shizhu Jin, Narayan Nepal, Yang Gao","doi":"10.1080/25785826.2021.1963190","DOIUrl":"https://doi.org/10.1080/25785826.2021.1963190","url":null,"abstract":"<p><p><i>Helicobacter pylori</i> (<i>HP</i>) is the primary etiologic factor that induces events in the immune system that lead to peptic ulcers. Toll-like receptors (TLRs) are an important part of the innate immune system, as they play pivotal roles in pathogen-associated molecular pattern (PAMP) recognition of <i>HP</i> as well host-associated damage-associated molecular patterns (DAMPs). Recent advancements such as COX-2 production, LPS recognition through TLR2, CagL, and CagY protein of <i>HP</i> activating TLR5, TLR9 activation via type IV secretion system (T4SS) using DNA transfer, TLR polymorphisms, their adaptor molecules, cytokines, and other factors play a significant role in PUD. Thus, some novel PUD treatments including Chuyou Yuyang granules, function by TLR4/NF-κB signaling pathway suppression and TNF-α and IL-18 inhibition also rely on TLR signaling. Similarly glycyrrhetinic acid (GA) treatment activates TLR-4 in Ana-1 cells not via TRIF, but via MYD88 expression, which is significantly upregulated to cure PUD. Therefore, understanding TLR signaling complexity and its resultant immune modulation after host-pathogen interactions is pivotal to drug and vaccine development for other diseases as well including cancer and recent pandemic COVID-19. In this review, we summarize the TLRs and <i>HP</i> interaction; its pathophysiology-related signaling pathways, polymorphisms, and pharmaceutical approaches toward PUD.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39525896","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01Epub Date: 2021-09-08DOI: 10.1080/25785826.2021.1975228
Agustinus Darmadi Hariyanto, Tiara Bunga Mayang Permata, Soehartati Argadikoesoema Gondhowiardjo
Not all T cells are effector cells of the anti-tumor immune system. One of the subpopulations of CD4+ T cells that express CD25+ and the transcription factor FOXP3, known as Regulator T cells (TReg), plays an essential role in maintaining tolerance and immune homeostasis preventing autoimmune diseases, minimalize chronic inflammatory diseases by enlisting various immunoregulatory mechanisms. The balance between effector T cells (Teff) and regulator T cells is crucial in determining the outcome of an immune response. Regarding tumors, activation or expansion of TReg cells reduces anti-tumor immunity. TReg cells inhibit the activation of CD4+ and CD8+ T cells and suppress anti-tumor activity in the tumor microenvironment. In addition, TReg cells also promote tumor angiogenesis both directly and indirectly to ensure oxygen and nutrient transport to the tumor. There is accumulating evidence showing a positive result that removing or suppressing TReg cells increases anti-tumor immune response. However, depletion of TReg cells will cause autoimmunity. One strategy to improve or restore tumor immunity is targeted therapy on the dominant effector TReg cells in tumor tissue. Various molecules such as CTLA-4, CD4, CD25, GITR, PD-1, OX40, ICOS are in clinical trials to assess their role in attenuating TReg cells' function.
{"title":"Role of CD4<sup>+</sup>CD25<sup>+</sup>FOXP3<sup>+</sup> T<sub>Reg</sub> cells on tumor immunity.","authors":"Agustinus Darmadi Hariyanto, Tiara Bunga Mayang Permata, Soehartati Argadikoesoema Gondhowiardjo","doi":"10.1080/25785826.2021.1975228","DOIUrl":"https://doi.org/10.1080/25785826.2021.1975228","url":null,"abstract":"<p><p>Not all T cells are effector cells of the anti-tumor immune system. One of the subpopulations of CD4<sup>+</sup> T cells that express CD25<sup>+</sup> and the transcription factor FOXP3, known as Regulator T cells (T<sub>Reg</sub>), plays an essential role in maintaining tolerance and immune homeostasis preventing autoimmune diseases, minimalize chronic inflammatory diseases by enlisting various immunoregulatory mechanisms. The balance between effector T cells (T<sub>eff</sub>) and regulator T cells is crucial in determining the outcome of an immune response. Regarding tumors, activation or expansion of T<sub>Reg</sub> cells reduces anti-tumor immunity. T<sub>Reg</sub> cells inhibit the activation of CD4<sup>+</sup> and CD8<sup>+</sup> T cells and suppress anti-tumor activity in the tumor microenvironment. In addition, T<sub>Reg</sub> cells also promote tumor angiogenesis both directly and indirectly to ensure oxygen and nutrient transport to the tumor. There is accumulating evidence showing a positive result that removing or suppressing T<sub>Reg</sub> cells increases anti-tumor immune response. However, depletion of T<sub>Reg</sub> cells will cause autoimmunity. One strategy to improve or restore tumor immunity is targeted therapy on the dominant effector T<sub>Reg</sub> cells in tumor tissue. Various molecules such as CTLA-4, CD4, CD25, GITR, PD-1, OX40, ICOS are in clinical trials to assess their role in attenuating T<sub>Reg</sub> cells' function.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39413323","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Despite the considerable success of cancer immunotherapy with immune checkpoint inhibitors, their nonspecific release of the immunosuppressive mechanism is often associated with immune-related adverse events (irAEs). irAEs significantly disturb patients' quality of life and can even be life-threatening. Therefore, the appropriate management of irAEs is crucial for the development of further reliable cancer immunotherapies. irAEs have the appearance of ordinary autoimmune diseases in one aspect but often have distinct features. Although the detailed pathogenesis of irAEs remains unclear, increasing numbers of studies have provided numerous clues. Here, we review the current knowledge on irAEs, particularly from an immunopathological basis.
{"title":"Immunopathological basis of immune-related adverse events induced by immune checkpoint blockade therapy.","authors":"Terufumi Kubo, Yoshihiko Hirohashi, Tomohide Tsukahara, Takayuki Kanaseki, Kenji Murata, Rena Morita, Toshihiko Torigoe","doi":"10.1080/25785826.2021.1976942","DOIUrl":"https://doi.org/10.1080/25785826.2021.1976942","url":null,"abstract":"<p><p>Despite the considerable success of cancer immunotherapy with immune checkpoint inhibitors, their nonspecific release of the immunosuppressive mechanism is often associated with immune-related adverse events (irAEs). irAEs significantly disturb patients' quality of life and can even be life-threatening. Therefore, the appropriate management of irAEs is crucial for the development of further reliable cancer immunotherapies. irAEs have the appearance of ordinary autoimmune diseases in one aspect but often have distinct features. Although the detailed pathogenesis of irAEs remains unclear, increasing numbers of studies have provided numerous clues. Here, we review the current knowledge on irAEs, particularly from an immunopathological basis.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39433164","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01Epub Date: 2021-09-01DOI: 10.1080/25785826.2021.1969116
Kayoko Kaneko, Nobuaki Ozawa, Atsuko Murashima
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by clinical manifestations such as thrombosis and obstetric complications with documented persistence of antiphospholipid antibodies (aPLs). Recent studies have revealed that the cause of aPL-related obstetric complications is dysfunction of placental trophoblasts and inflammation of the maternal-fetal interface induced by aPLs, not thrombosis. Although aPLs are associated with recurrence of serious complications during pregnancy, appropriate combination therapy with heparin and low-dose aspirin can improve the course of 70-80% of subsequent pregnancies. Preconception counseling and patient-tailored treatment are fundamental to improving maternal and fetal outcomes. Non-anticoagulant treatments such as hydroxychloroquine and statins are being developed for cases refractory to conventional treatment. Risk factors for thrombosis after pregnancy complications were identified based on the analysis of large databases of obstetric APS.
{"title":"Obstetric anti-phospholipid syndrome: from pathogenesis to treatment.","authors":"Kayoko Kaneko, Nobuaki Ozawa, Atsuko Murashima","doi":"10.1080/25785826.2021.1969116","DOIUrl":"https://doi.org/10.1080/25785826.2021.1969116","url":null,"abstract":"<p><p>Antiphospholipid syndrome (APS) is an autoimmune disease characterized by clinical manifestations such as thrombosis and obstetric complications with documented persistence of antiphospholipid antibodies (aPLs). Recent studies have revealed that the cause of aPL-related obstetric complications is dysfunction of placental trophoblasts and inflammation of the maternal-fetal interface induced by aPLs, not thrombosis. Although aPLs are associated with recurrence of serious complications during pregnancy, appropriate combination therapy with heparin and low-dose aspirin can improve the course of 70-80% of subsequent pregnancies. Preconception counseling and patient-tailored treatment are fundamental to improving maternal and fetal outcomes. Non-anticoagulant treatments such as hydroxychloroquine and statins are being developed for cases refractory to conventional treatment. Risk factors for thrombosis after pregnancy complications were identified based on the analysis of large databases of obstetric APS.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39375168","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01Epub Date: 2022-02-07DOI: 10.1080/25785826.2022.2031812
Bo-Young Shin, Su-Hyeon Lee, Yuna Kim, Jaekyeung An, Tae-Yoon Park, Sang-Kyou Lee
Th17 cells are implicated in the pathogenesis of several autoimmune diseases. During the inflammation, Th17 cells exposed to IL-12 can shift towards the Th1 phenotype. These shifted cells are defined as 'non-classic Th1 cells'. Th17-derived non-classic Th1 cells play a critical role in late-onset chronic inflammatory diseases and are more pathogenic than the unshifted Th17 cells. Eomes is a transcription factor highly expressed in non-classic Th1 cells. To study the functional role of Eomes without genetic alteration, novel recombinant protein, ntEomes-TMD, was generated by fusing TMD of Eomes and Hph-1-PTD that facilitate intracellular delivery of its cargo molecule. ntEomes-TMD was delivered into the nucleus of the cells without influencing the T cell activation and cytotoxicity. ntEomes-TMD specifically inhibited the Eomes- and ROR-γt-mediated transcription and suppressed the Th1 and Th17 differentiation. Interestingly, ntEomes-TMD blocked the generation of non-classic Th1 cells from Th17 cells, leading to the inhibition of IFN-γ and GM-CSF secretion. In EAE, ntEomes-TMD alleviated the symptoms of EAE, and the combination treatment using ntEomes-TMD and anti-IL-17 mAb together showed better therapeutic efficacy than anti-IL-17 mAb treatment. The results suggest that ntEomes-TMD can be a new therapeutic reagent for treating chronic inflammatory diseases associated with non-classic Th1 cells.
{"title":"Interactomic inhibition of Eomes in the nucleus alleviates EAE via blocking the conversion of Th17 cells into non-classic Th1 cells.","authors":"Bo-Young Shin, Su-Hyeon Lee, Yuna Kim, Jaekyeung An, Tae-Yoon Park, Sang-Kyou Lee","doi":"10.1080/25785826.2022.2031812","DOIUrl":"https://doi.org/10.1080/25785826.2022.2031812","url":null,"abstract":"<p><p>Th17 cells are implicated in the pathogenesis of several autoimmune diseases. During the inflammation, Th17 cells exposed to IL-12 can shift towards the Th1 phenotype. These shifted cells are defined as 'non-classic Th1 cells'. Th17-derived non-classic Th1 cells play a critical role in late-onset chronic inflammatory diseases and are more pathogenic than the unshifted Th17 cells. Eomes is a transcription factor highly expressed in non-classic Th1 cells. To study the functional role of Eomes without genetic alteration, novel recombinant protein, ntEomes-TMD, was generated by fusing TMD of Eomes and Hph-1-PTD that facilitate intracellular delivery of its cargo molecule. ntEomes-TMD was delivered into the nucleus of the cells without influencing the T cell activation and cytotoxicity. ntEomes-TMD specifically inhibited the Eomes- and ROR-γt-mediated transcription and suppressed the Th1 and Th17 differentiation. Interestingly, ntEomes-TMD blocked the generation of non-classic Th1 cells from Th17 cells, leading to the inhibition of IFN-γ and GM-CSF secretion. In EAE, ntEomes-TMD alleviated the symptoms of EAE, and the combination treatment using ntEomes-TMD and anti-IL-17 mAb together showed better therapeutic efficacy than anti-IL-17 mAb treatment. The results suggest that ntEomes-TMD can be a new therapeutic reagent for treating chronic inflammatory diseases associated with non-classic Th1 cells.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39600508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01Epub Date: 2021-08-15DOI: 10.1080/25785826.2021.1963189
Jun Miyoshi, Tadakazu Hisamatsu
Antibiotics are widely prescribed for mothers in the peripartum period today. Approximately 40% of pregnant women at term are exposed to antibiotics. Antibiotics are useful against infectious conditions such as chorioamnionitis; however, they alter the maternal microbiome. The maternal microbiome, particularly the gut microbiome, is transmitted to their neonates and is one of the major sources that shape the child's gut microbiome. The gut microbiome early in life plays a crucial role in the development of the gut microbiome itself as well as the host health over the entire life. Microbes structure the commensal ecosystem in the host. Simultaneously, microbial components and metabolites influence the host organ functions including the immune system, and vice versa, the various factors of the host impact the microbiome. The alterations of the gut microbiome induced by antibiotics in mothers can lead to gut dysbiosis in children eventually resulting in chronic disease conditions including immune disorders. Knowledge of the lasting impacts of maternal peripartum exposure to antibiotics on the gut microbiome and health in offspring and reconsideration of the adequate use of antibiotics in clinical practice are needed. Avoiding and restoring neonatal dysbiosis following maternal antibiotics-induced dysbiosis could be a new preventive strategy for various diseases.
{"title":"The impact of maternal exposure to antibiotics on the development of child gut microbiome.","authors":"Jun Miyoshi, Tadakazu Hisamatsu","doi":"10.1080/25785826.2021.1963189","DOIUrl":"https://doi.org/10.1080/25785826.2021.1963189","url":null,"abstract":"<p><p>Antibiotics are widely prescribed for mothers in the peripartum period today. Approximately 40% of pregnant women at term are exposed to antibiotics. Antibiotics are useful against infectious conditions such as chorioamnionitis; however, they alter the maternal microbiome. The maternal microbiome, particularly the gut microbiome, is transmitted to their neonates and is one of the major sources that shape the child's gut microbiome. The gut microbiome early in life plays a crucial role in the development of the gut microbiome itself as well as the host health over the entire life. Microbes structure the commensal ecosystem in the host. Simultaneously, microbial components and metabolites influence the host organ functions including the immune system, and vice versa, the various factors of the host impact the microbiome. The alterations of the gut microbiome induced by antibiotics in mothers can lead to gut dysbiosis in children eventually resulting in chronic disease conditions including immune disorders. Knowledge of the lasting impacts of maternal peripartum exposure to antibiotics on the gut microbiome and health in offspring and reconsideration of the adequate use of antibiotics in clinical practice are needed. Avoiding and restoring neonatal dysbiosis following maternal antibiotics-induced dysbiosis could be a new preventive strategy for various diseases.</p>","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39322977","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-05-02DOI: 10.1080/25785826.2022.2066251
Dennis Jiménez, Marbel Torres Arias
Abstract SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19. Graphical abstract
{"title":"Immunouniverse of SARS-CoV-2","authors":"Dennis Jiménez, Marbel Torres Arias","doi":"10.1080/25785826.2022.2066251","DOIUrl":"https://doi.org/10.1080/25785826.2022.2066251","url":null,"abstract":"Abstract SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19. Graphical abstract","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41616258","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-05-01DOI: 10.1080/25785826.2022.2068331
Neelam Asghar, H. Mumtaz, Abdul Ahad Syed, Farea Eqbal, Reeju Maharjan, Aditya Bamboria, Manish Shrehta
Abstract The World Health Organization stated on 11 March 2020 that a coronavirus illness had been discovered in Wuhan, China in December 2019. Effective vaccinations are eagerly awaited as the global outbreak of COVID-19 continues. The aim is to evaluate the safety, effectiveness, and immunogenicity of Pfizer/AstraZeneca/Modera/Cansino vaccines against COVID-19. An electronic search on different databases yielded 12,907 articles. A total of 20 randomized and non-randomized, published, and ongoing trials were selected. Cochrane RoB version 2.0 was used to assess the authenticity of the studies. Of these 20 trials, three were conducted on Pfizer, three on AstraZeneca, three on Moderna, and two on the Cansino vaccine. These trials have reported promising results for the safety, efficacy, and immunogenicity of the respective vaccines. None of the trials have reported the efficacy and severe adverse outcomes for the Cansino vaccine, hindering its reliability as a safe vaccine against covid-19. Furthermore, the results of these trials have established Pfizer to be the most efficacious vaccine against covid-19, having an efficacy of 94.6%. A few severe adverse events were reported by the included trials. However, further systematic reviews are required to understand the respective vaccine profiles on Immuno-suppressive, organ transplants, and patients with other comorbidities.
摘要世界卫生组织于2020年3月11日表示,2019年12月在中国武汉发现了一种冠状病毒疾病。随着新冠肺炎全球疫情的持续,人们热切期待有效的疫苗接种。目的是评估辉瑞/阿斯利康/莫德纳/康西诺疫苗对抗新冠肺炎的安全性、有效性和免疫原性。对不同数据库进行的电子搜索共产生12907篇文章。共选择了20项随机和非随机、已发表和正在进行的试验。Cochrane RoB 2.0版用于评估研究的真实性。在这20项试验中,三项是在辉瑞公司进行的,三项在阿斯利康公司进行的、三项在莫德纳公司进行的和两项在康西诺疫苗上进行的。这些试验报告了各自疫苗的安全性、有效性和免疫原性方面有希望的结果。没有一项试验报告了Cansino疫苗的有效性和严重不良后果,这阻碍了其作为新冠肺炎安全疫苗的可靠性。此外,这些试验的结果表明,辉瑞公司是最有效的新冠肺炎疫苗,其有效性为94.6%。纳入的试验报告了一些严重不良事件。然而,需要进一步的系统审查,以了解免疫抑制、器官移植和其他合并症患者各自的疫苗概况。
{"title":"Safety, efficacy, and immunogenicity of COVID-19 vaccines; a systematic review","authors":"Neelam Asghar, H. Mumtaz, Abdul Ahad Syed, Farea Eqbal, Reeju Maharjan, Aditya Bamboria, Manish Shrehta","doi":"10.1080/25785826.2022.2068331","DOIUrl":"https://doi.org/10.1080/25785826.2022.2068331","url":null,"abstract":"Abstract The World Health Organization stated on 11 March 2020 that a coronavirus illness had been discovered in Wuhan, China in December 2019. Effective vaccinations are eagerly awaited as the global outbreak of COVID-19 continues. The aim is to evaluate the safety, effectiveness, and immunogenicity of Pfizer/AstraZeneca/Modera/Cansino vaccines against COVID-19. An electronic search on different databases yielded 12,907 articles. A total of 20 randomized and non-randomized, published, and ongoing trials were selected. Cochrane RoB version 2.0 was used to assess the authenticity of the studies. Of these 20 trials, three were conducted on Pfizer, three on AstraZeneca, three on Moderna, and two on the Cansino vaccine. These trials have reported promising results for the safety, efficacy, and immunogenicity of the respective vaccines. None of the trials have reported the efficacy and severe adverse outcomes for the Cansino vaccine, hindering its reliability as a safe vaccine against covid-19. Furthermore, the results of these trials have established Pfizer to be the most efficacious vaccine against covid-19, having an efficacy of 94.6%. A few severe adverse events were reported by the included trials. However, further systematic reviews are required to understand the respective vaccine profiles on Immuno-suppressive, organ transplants, and patients with other comorbidities.","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43565304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-04-07DOI: 10.1080/25785826.2022.2060169
T. Shimada, M. Higashida-Konishi, M. Akiyama, S. Hama, K. Izumi, S. Matsubara, H. Oshima, Y. Okano
Abstract Currently, no standard treatment strategy has been established for immune-mediated necrotizing myopathy (IMNM). Here we present a case of IMNM which was successfully treated with intensive combined therapy with high-dose glucocorticoids, tacrolimus, and intravenous immunoglobulins. Her muscle weakness was rapidly progressive and severe so that she became bedridden one week after admission. She was complicated with dysphagia and had serum myogenic enzymes elevation, ventricular diastolic dysfunction, and interstitial lung disease. Serum anti-SRP antibody was positive and her muscle biopsy revealed many necrotic fibers with minimal inflammation. Further histological analysis demonstrated infiltration of phagocytic macrophages with deposition of membrane attack complex (C5b-9) in the necrotic muscle fibers, suggesting activation of complement pathway and macrophages as a pathomechanism of this disease. She was diagnosed as IMNM and was immediately initiated a combination therapy described above, which led to dramatic clinical improvements. Recent studies suggest that intravenous immunoglobulins and tacrolimus can inhibit the activation of complement pathway and macrophages. Our present case suggests that early initiation of intensive combined therapy including intravenous immunoglobulins and tacrolimus might be effective for preventing irreversible muscle damages by disrupting a pathogenic activation of complement and macrophages in IMNM.
{"title":"Immune-mediated necrotizing myopathy which showed deposition of C5b-9 in the necrotic muscle fibers and was successfully treated with intensive combined therapy with high-dose glucocorticoids, tacrolimus, and intravenous immunoglobulins","authors":"T. Shimada, M. Higashida-Konishi, M. Akiyama, S. Hama, K. Izumi, S. Matsubara, H. Oshima, Y. Okano","doi":"10.1080/25785826.2022.2060169","DOIUrl":"https://doi.org/10.1080/25785826.2022.2060169","url":null,"abstract":"Abstract Currently, no standard treatment strategy has been established for immune-mediated necrotizing myopathy (IMNM). Here we present a case of IMNM which was successfully treated with intensive combined therapy with high-dose glucocorticoids, tacrolimus, and intravenous immunoglobulins. Her muscle weakness was rapidly progressive and severe so that she became bedridden one week after admission. She was complicated with dysphagia and had serum myogenic enzymes elevation, ventricular diastolic dysfunction, and interstitial lung disease. Serum anti-SRP antibody was positive and her muscle biopsy revealed many necrotic fibers with minimal inflammation. Further histological analysis demonstrated infiltration of phagocytic macrophages with deposition of membrane attack complex (C5b-9) in the necrotic muscle fibers, suggesting activation of complement pathway and macrophages as a pathomechanism of this disease. She was diagnosed as IMNM and was immediately initiated a combination therapy described above, which led to dramatic clinical improvements. Recent studies suggest that intravenous immunoglobulins and tacrolimus can inhibit the activation of complement pathway and macrophages. Our present case suggests that early initiation of intensive combined therapy including intravenous immunoglobulins and tacrolimus might be effective for preventing irreversible muscle damages by disrupting a pathogenic activation of complement and macrophages in IMNM.","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42211111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-04-06DOI: 10.1080/25785826.2022.2060168
Adrian Y. S. Lee
Abstract Anti-histone antibodies (AHAs) make their appearance in a number of systemic autoimmune diseases including systemic lupus erythematosus (SLE) and drug-induced lupus erythematosus (DILE). Although being known for over 50 years, they are poorly studied and understood. There is emerging evidence for their use in predicting clinical features of SLE, diversifying their clinical use. AHAs, however, are probably less prevalent in DILE than once thought owing to a move away from older DILE drugs to modern biological agents which do not appear to elicit AHAs. This review examines the historical studies that have defined AHAs and looks at some of the recent work with these autoantibodies.
{"title":"Clinical use of anti-histone antibodies in idiopathic and drug-induced lupus","authors":"Adrian Y. S. Lee","doi":"10.1080/25785826.2022.2060168","DOIUrl":"https://doi.org/10.1080/25785826.2022.2060168","url":null,"abstract":"Abstract Anti-histone antibodies (AHAs) make their appearance in a number of systemic autoimmune diseases including systemic lupus erythematosus (SLE) and drug-induced lupus erythematosus (DILE). Although being known for over 50 years, they are poorly studied and understood. There is emerging evidence for their use in predicting clinical features of SLE, diversifying their clinical use. AHAs, however, are probably less prevalent in DILE than once thought owing to a move away from older DILE drugs to modern biological agents which do not appear to elicit AHAs. This review examines the historical studies that have defined AHAs and looks at some of the recent work with these autoantibodies.","PeriodicalId":37286,"journal":{"name":"Immunological Medicine","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2022-04-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46054589","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}