Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.05.003
Syed Shahzad Hasan , Abdur Rashid , Sundus Osama , Zia Ul Mustafa , Hamid A. Merchant
Covid immunization commenced on 2nd Feb 2021 in Pakistan and as of 7th Sep 2021, over 84 million vaccine doses were administered in Pakistan, of which 72% procured by the government, 22% received through Covax and 6% were donated. The vaccines rolled out nationally included: Sinopharm, Sinovac and CanSinoBIO (China), AstraZeneca (UK), Moderna and Pfizer (USA), Sputnik (Russia), and PakVac (China/Pakistan). About half of the eligible population in Pakistan (63 m) had received at least one dose of Covid vaccine as of Sep 2021. Pakistan National Pharmacovigilance Centre (PNPC) in coordination with WHO, MHRA and Uppsala Monitoring Centre (UMC) established pharmacovigilance centers across Pakistan. The Covid vaccine AEFIs in Pakistan were mainly reported via NIMS (National Immunization Management System), COVIM (Covid-19 Vaccine Inventory Management System), 1166 freephone helpline and MedSafety. There have been 39,291 ADRs reported as of 30th Sept 2021, where most reported after the first dose (n = 27,108) and within 24–72 h of immunization (n = 27,591). Fever or shivering accounted for most AEFI (35%) followed by injection-site pain or redness (28%), headache (26%), nausea/vomiting (4%), and diarrhoea (3%). 24 serious AEFIs were also reported and investigated in detail by the National AEFI review committee. The rate of AEFIs reports ranged from 0.27 to 0.79 per 1000 for various Covid vaccines in Pakistan that was significantly lower than the rates in UK (∼4 per 1000), primarily atrributed to underreporting of cases in Pakistan. Finally, Covid vaccines were well tolerated and no significant cause for concern was flagged up in Pakistan's Covid vaccine surveillance system concluding overall benefits outweighed risks.
{"title":"Covid-19 Vaccine safety and adverse event analysis from Pakistan","authors":"Syed Shahzad Hasan , Abdur Rashid , Sundus Osama , Zia Ul Mustafa , Hamid A. Merchant","doi":"10.1016/j.clicom.2022.05.003","DOIUrl":"10.1016/j.clicom.2022.05.003","url":null,"abstract":"<div><p>Covid immunization commenced on 2nd Feb 2021 in Pakistan and as of 7th Sep 2021, over 84 million vaccine doses were administered in Pakistan, of which 72% procured by the government, 22% received through Covax and 6% were donated. The vaccines rolled out nationally included: Sinopharm, Sinovac and CanSinoBIO (China), AstraZeneca (UK), Moderna and Pfizer (USA), Sputnik (Russia), and PakVac (China/Pakistan). About half of the eligible population in Pakistan (63 m) had received at least one dose of Covid vaccine as of Sep 2021. Pakistan National Pharmacovigilance Centre (PNPC) in coordination with WHO, MHRA and Uppsala Monitoring Centre (UMC) established pharmacovigilance centers across Pakistan. The Covid vaccine AEFIs in Pakistan were mainly reported via NIMS (National Immunization Management System), COVIM (Covid-19 Vaccine Inventory Management System), 1166 freephone helpline and MedSafety. There have been 39,291 ADRs reported as of 30th Sept 2021, where most reported after the first dose (<em>n</em> = 27,108) and within 24–72 h of immunization (<em>n</em> = 27,591). Fever or shivering accounted for most AEFI (35%) followed by injection-site pain or redness (28%), headache (26%), nausea/vomiting (4%), and diarrhoea (3%). 24 serious AEFIs were also reported and investigated in detail by the National AEFI review committee. The rate of AEFIs reports ranged from 0.27 to 0.79 per 1000 for various Covid vaccines in Pakistan that was significantly lower than the rates in UK (∼4 per 1000), primarily atrributed to underreporting of cases in Pakistan. Finally, Covid vaccines were well tolerated and no significant cause for concern was flagged up in Pakistan's Covid vaccine surveillance system concluding overall benefits outweighed risks.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000154/pdfft?md5=c5f792b4c679597f1d020f0a7e3929ef&pid=1-s2.0-S2772613422000154-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74011765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
We conducted an experimental protocol with a nasal recombinant IFNα-2b formulation (Nasalferon) in 12 healthy volunteers who received 1 MIU per nostril twice a day, three consecutive days, and studied the induction of biomarkers related to antiviral and immune responses. Nasalferon increases the antiviral biomarker OAS1 transcript levels in oropharynx and PBMCs, regulates molecular and cellular elements related to innate and adaptive immune responses and decreases granulocytes population. These effects support Nasalferon use in virally-exposed populations.
{"title":"Nasalferon, a new nasal formulation of IFNα2b, modulates cellular and molecular elements associated with an antiviral response in mucosa and blood","authors":"Dania Vázquez-Blomquist , Mónica Bequet-Romero , Yssel Mendoza-Marí , Gilda Lemos , Iris Valdés , Camila Canaán-Haden Ayala , Isabel Gonzáles Moya , Ricardo Martínez Rosales , Giselle Freyre Corrales , Gerardo Guillén-Nieto","doi":"10.1016/j.clicom.2022.02.002","DOIUrl":"10.1016/j.clicom.2022.02.002","url":null,"abstract":"<div><p>We conducted an experimental protocol with a nasal recombinant IFNα-2b formulation (Nasalferon) in 12 healthy volunteers who received 1 MIU per nostril twice a day, three consecutive days, and studied the induction of biomarkers related to antiviral and immune responses. Nasalferon increases the antiviral biomarker OAS1 transcript levels in oropharynx and PBMCs, regulates molecular and cellular elements related to innate and adaptive immune responses and decreases granulocytes population. These effects support Nasalferon use in virally-exposed populations.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000051/pdfft?md5=92213432cd7e3694dd11ab0804a3bb79&pid=1-s2.0-S2772613422000051-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79646466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The role of thymic stromal lymphopoietin (TSLP) in chronic infection is not well understood. We report an increased number of cluster of differentiation 8 (CD8+ T) cells and higher expression of TSLP in the tonsil tissues of patients with recurrent tonsillitis as compared to that in the tonsil tissues of patients with tonsillar hypertrophy. Stimulation of tonsil CD8+ T cells with TSLP in the recurrent tonsillitis group resulted in higher production of interferon gamma (IFN-γ) and increased cell proliferation when compared to that in the tonsillar hypertrophy group. These results suggest the possibility that recurrent infection of tonsils may enhance local CD8+ T cell proliferation and IFN-γ and TSLP production. Anti-TSLP antibodies, which are potential therapeutic agents in allergic diseases, could have beneficial effects in patients with recurrent tonsillitis.
{"title":"TSLP activates the production of IFN-γ via CD8-positive T cells in recurrent tonsillitis","authors":"Kota Hiraoka, Toshiaki Kawano, Takashi Hirano, Masashi Urabe, Kaori Tateyama, Masashi Suzuki","doi":"10.1016/j.clicom.2022.05.002","DOIUrl":"10.1016/j.clicom.2022.05.002","url":null,"abstract":"<div><p>The role of thymic stromal lymphopoietin (TSLP) in chronic infection is not well understood. We report an increased number of cluster of differentiation 8 (CD8+ T) cells and higher expression of TSLP in the tonsil tissues of patients with recurrent tonsillitis as compared to that in the tonsil tissues of patients with tonsillar hypertrophy. Stimulation of tonsil CD8+ T cells with TSLP in the recurrent tonsillitis group resulted in higher production of interferon gamma (IFN-γ) and increased cell proliferation when compared to that in the tonsillar hypertrophy group. These results suggest the possibility that recurrent infection of tonsils may enhance local CD8+ T cell proliferation and IFN-γ and TSLP production. Anti-TSLP antibodies, which are potential therapeutic agents in allergic diseases, could have beneficial effects in patients with recurrent tonsillitis.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000142/pdfft?md5=211611bc8be715a6c161329c2208c338&pid=1-s2.0-S2772613422000142-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89400149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.03.005
M D Ray , SVS Deo , Kalpana Luthra , Sandeep Mathur , Prem Anand , Romsha Kumar , Shaifali Sharma , Shruti Kahol , Abdul Wahid
Background
Despite of multimodality treatment of Ovarian Cancer (OC), about two-thirds of patients have peritoneal recurrence with 5-year survival of 15–35%. HIPEC (Hyperthermic Intraperitoneal Chemotherapy) delivers cell-cycle non-specific chemotherapeutic agents along with the temperature of 41°C to 43 °C into the peritoneal cavity after achieving complete cytoreduction, improving overall survival around 50% and recurrence-free survival significantly. Cytoreductive surgery (CRS) and HIPEC may be UpfrontOR Primary - when performed primarily, Interval - when performed after 3 to 6 six cycles of Neo-adjuvant chemotherapy(NACT) and Secondary-when performed for patients who have a recurrence after CRS. OC is associated frequently with tumor-infiltrating lymphocytes in OC microenvironment. Here, we sought to study the interaction of HIPEC on FOXp3 and RORɣ T cells in primary, interval and recurrent settings of HIPEC.
Methodology
We have seen the expression of FOXp3, RORɣ in tumor tissue and surrounding normal tissue of ovarian cancer patients. We also performed IHC of tumor tissue and qRT-PCR of cDNA, isolated from tumor tissue and adjacent normal tissue for the same expression and compared intra-group and among the groups.We studied the expression of FOXp3,RORɣ, Interleukin-10 (IL-10) and Interleukin-17 (IL-17) in peripheral blood mononuclear cells(PBMC) before and after HIPEC in one week and four weeks and compared the changes of expressions. We included 30 patients with FIGO stage III to IVA advanced Epithelial ovarian cancer patients, with minimum 10 patients in each subgroup i.e.,upfront, interval and secondary. The pilot study was conducted between June 2017 to February 2021 at the premier Institute, India after Ethical approval.
Results
Though there was highest expression of FOXp3 mRNA in recurrent group followed by upfront and interval. However, there was no difference in RORɣ T cell mRNA (by qRT-PCR) expression among three subgroups. We found the change in the expression of FOXp3 and RORɣ T cell in Peripheral blood mononuclear cells by flowcytometry, before and after HIPEC (around 1 week and 4 week after HIPEC) to be significant. In the interval group, there was decreased expression of pre-op Foxp3 compared to the upfront (p = 0.0121) and control (p = 0.0187). These findings suggest that neoadjuvant chemotherapy may reverse the immunosuppressive environment in ovarian cancers. In the upfront group, there was a transient increase of FOXp3 in one week post HIPEC (p = 0.005) and then it decreased to below pre-op values in 4 weeks (p = 0.022). There was no difference in pre-op RORγ and IL 17 among the three subgroups.However, RORγ was increased at four weeks post-HIPEC in the upfront group (p = 0.009), interval group (p = 0.032), and the recurrent group. (
{"title":"Molecular linking of HIPEC (Hyperthermic Intraperitoneal Chemotherapy) and T-regulatory cells (FOXp3) and Th-17 (RORɣ) in advanced epithelial ovarian cancer","authors":"M D Ray , SVS Deo , Kalpana Luthra , Sandeep Mathur , Prem Anand , Romsha Kumar , Shaifali Sharma , Shruti Kahol , Abdul Wahid","doi":"10.1016/j.clicom.2022.03.005","DOIUrl":"10.1016/j.clicom.2022.03.005","url":null,"abstract":"<div><h3>Background</h3><p>Despite of multimodality treatment of Ovarian Cancer (OC), about two-thirds of patients have peritoneal recurrence with 5-year survival of 15–35%. HIPEC (Hyperthermic Intraperitoneal Chemotherapy) delivers cell-cycle non-specific chemotherapeutic agents along with the temperature of 41°C to 43 °C into the peritoneal cavity after achieving complete cytoreduction, improving overall survival around 50% and recurrence-free survival significantly. Cytoreductive surgery (CRS) and HIPEC may be <strong>Upfront</strong>OR <strong>Primary -</strong> when performed primarily<strong>, Interval -</strong> when performed after 3 to 6 six cycles of Neo-adjuvant chemotherapy(NACT) and S<strong>econdary-</strong>when performed for patients who have a recurrence after CRS. OC is associated frequently with tumor-infiltrating lymphocytes in OC microenvironment. Here, we sought to study the interaction of HIPEC on FOXp3 and RORɣ T cells in primary, interval and recurrent settings of HIPEC.</p></div><div><h3>Methodology</h3><p>We have seen the expression of FOXp3, RORɣ in tumor tissue and surrounding normal tissue of ovarian cancer patients. We also performed IHC of tumor tissue and qRT-PCR of cDNA, isolated from tumor tissue and adjacent normal tissue for the same expression and compared intra-group and among the groups.We studied the expression of FOXp3,RORɣ, Interleukin-10 (IL-10) and Interleukin-17 (IL-17) in peripheral blood mononuclear cells(PBMC) before and after HIPEC in one week and four weeks and compared the changes of expressions. We included 30 patients with FIGO stage III to IVA advanced Epithelial ovarian cancer patients, with minimum 10 patients in each subgroup i.e.,upfront, interval and secondary. The pilot study was conducted between June 2017 to February 2021 at the premier Institute, India after Ethical approval.</p></div><div><h3>Results</h3><p>Though there was highest expression of FOXp3 mRNA in recurrent group followed by upfront and interval. However, there was no difference in RORɣ T cell mRNA (by qRT-PCR) expression among three subgroups. We found the change in the expression of FOXp3 and RORɣ T cell in Peripheral blood mononuclear cells by flowcytometry, before and after HIPEC (around 1 week and 4 week after HIPEC) to be significant. In the interval group, there was decreased expression of pre-op Foxp3 compared to the upfront (<em>p</em> = 0.0121) and control (<em>p</em> = 0.0187). These findings suggest that neoadjuvant chemotherapy may reverse the immunosuppressive environment in ovarian cancers. In the upfront group, there was a transient increase of FOXp3 in one week post HIPEC (<em>p</em> = 0.005) and then it decreased to below pre-op values in 4 weeks (<em>p</em> = 0.022). There was no difference in pre-op RORγ and IL 17 among the three subgroups.However, RORγ was increased at four weeks post-HIPEC in the upfront group (<em>p</em> = 0.009), interval group (<em>p</em> = 0.032), and the recurrent group. (<em>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000117/pdfft?md5=8e67252b7768ad788b550654bd098c02&pid=1-s2.0-S2772613422000117-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80635623","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.08.004
Suhana Mishra , Amir Mohammad Arsh , Jitendra Singh Rathore
Emerging research shows that innate immunity can also keep the memory of prior experiences, challenging the long-held notion that immunological memory is only the domain of the adaptive immune cells. However, the absence of immunological memory in innate immune responses has recently been brought into question. Now it is known that after a few transient activations, innate immune cells may acquire immunological memory phenotype, resulting in a stronger response to a subsequent secondary challenge. When exposed to particular microbial and/or inflammatory stimuli, trained innate immunity is characterized by the enhanced non-specific response, which is regulated by substantial metabolic alterations and epigenetic reprogramming. Trained immunity is acquired by two main reprogramming, namely, epigenetic reprogramming and metabolic adaptation/reprogramming. Epigenetic reprogramming causes changes in gene expression and cell physiology, resulting in internal cell signaling and/or accelerated and amplified cytokine release. Metabolic changes due to trained immunity induce accelerated glycolysis and glutaminolysis. As a result, trained immunity can have unfavorable outcomes, such as hyper inflammation and the development of cardiovascular diseases, autoinflammatory diseases, and neuroinflammation. In this review, the current scenario in the area of trained innate immunity, its mechanisms, and its involvement in immunological disorders are briefly outlined.
{"title":"Trained innate immunity and diseases: Bane with the boon","authors":"Suhana Mishra , Amir Mohammad Arsh , Jitendra Singh Rathore","doi":"10.1016/j.clicom.2022.08.004","DOIUrl":"10.1016/j.clicom.2022.08.004","url":null,"abstract":"<div><p>Emerging research shows that innate immunity can also keep the memory of prior experiences, challenging the long-held notion that immunological memory is only the domain of the adaptive immune cells. However, the absence of immunological memory in innate immune responses has recently been brought into question. Now it is known that after a few transient activations, innate immune cells may acquire immunological memory phenotype, resulting in a stronger response to a subsequent secondary challenge. When exposed to particular microbial and/or inflammatory stimuli, trained innate immunity is characterized by the enhanced non-specific response, which is regulated by substantial metabolic alterations and epigenetic reprogramming. Trained immunity is acquired by two main reprogramming, namely, epigenetic reprogramming and metabolic adaptation/reprogramming. Epigenetic reprogramming causes changes in gene expression and cell physiology, resulting in internal cell signaling and/or accelerated and amplified cytokine release. Metabolic changes due to trained immunity induce accelerated glycolysis and glutaminolysis. As a result, trained immunity can have unfavorable outcomes, such as hyper inflammation and the development of cardiovascular diseases, autoinflammatory diseases, and neuroinflammation. In this review, the current scenario in the area of trained innate immunity, its mechanisms, and its involvement in immunological disorders are briefly outlined.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000208/pdfft?md5=bc92de732b3ff9c163ed4ad5cb40b59f&pid=1-s2.0-S2772613422000208-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83222063","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.02.001
Pauline Veyrard , Xavier Roblin , Céline Pansart , Ren Mao , Stéphane Nancey , Martin Killian , Louis Waeckel , Anne-Emmanuelle Berger , Nicolas Williet , Laetitia Bastide , Mathilde Barrau , Quentin Tournier , Stéphane Paul
Aim
Serum calprotectin (SC), a novel biomarker of inflammatory bowel diseases (IBD), has been recently investigated with conflicting results. The purpose of this study was to assess the ability of SC to predict relapse in IBD patients treated by biologic therapies, and to evaluate the correlation between SC, clinical and endoscopic relapse and other biomarkers as fecal calprotectin (FC) and C-reactive protein (CRP).
Methods
All consecutive IBD patients in deep remission (clinical, endoscopic or imaging remission) were followed 12 months in this prospective study. Blood and stool samples were collected for SC, serum CRP and FC. SC was measured the day of inclusion (baseline, D0), 3 months (M3) and at 6 months (M6) or during the study period for clinical relapse. Relapse was defined as clinical, biomarkers, or endoscopic/imaging activities. Evolution of SC was quantified before relapse to analyze a predicting value of loss of response (LOR). SC for patients with active IBD and those with symptoms without inflammation were also compared.
Results
Among the 119 patients included, 54 (46.4%) patients experienced a disease relapse during follow-up. Median SC levels did not increase in patients with clinical relapse (3.15 µg/ml at baseline, 3.38 µg/ml at M3, 3.33 µg/ml at M6 and 3.99 µg/ml in case of relapse (p = 0.63)). SC were compared during relapse in patients with endoscopic remission but clinical symptoms defined as secondary Irritable Bowel Syndrome (IBS). SC levels were higher in active IBD and similar between the groups of patients with IBS or deep remission (3.05 µg/ml IBS vs 2.99 µg/ml remission vs 5.1 µg/ml for clinical relapse, p = 0.04). In patients with clinical symptoms, SC presents a good predictive value for relapse (AUROC 0.764, IC95: 0.68–0.88), with a sensitivity of 72%, a specificity of 77%, using a cut-off value of 4.45 µg/ml. A weak, but significant correlation was found between SC and FC levels (r = 0.35, P = 0.001). A combined score with CRP, FC and SC is not efficient to improve IBD diagnostic.
Conclusion
SC was significantly higher in patients with clinical relapse compared to those with endoscopic remission with or without clinical symptoms. SC allow to discriminate patients with active IBD or with IBS but failed to predict relapse.
血清钙保护蛋白(SC)是炎症性肠病(IBD)的一种新的生物标志物,最近的研究结果相互矛盾。本研究的目的是评估SC预测IBD生物治疗患者复发的能力,并评估SC、临床和内镜下复发以及其他生物标志物如粪便钙保护蛋白(FC)和c反应蛋白(CRP)之间的相关性。方法在这项前瞻性研究中,所有IBD深度缓解(临床、内镜或影像学缓解)患者连续随访12个月。采集血液和粪便标本检测SC、血清CRP和FC。SC在纳入当日(基线,D0)、3个月(M3)、6个月(M6)或研究期间临床复发时测量。复发定义为临床、生物标志物或内窥镜/成像活动。在复发前量化SC的演变,以分析反应丧失(LOR)的预测值。还比较了活动性IBD患者和无炎症症状患者的SC。结果119例患者中,54例(46.4%)患者在随访期间出现疾病复发。临床复发患者中位SC水平没有增加(基线为3.15µg/ml, M3为3.38µg/ml, M6为3.33µg/ml,复发时为3.99µg/ml (p = 0.63))。比较内镜下缓解但临床症状定义为继发性肠易激综合征(IBS)的患者复发期间的SC。SC水平在活动性IBD中较高,在IBS或深度缓解患者组之间相似(IBS组为3.05µg/ml vs缓解组为2.99µg/ml vs临床复发组为5.1µg/ml, p = 0.04)。在有临床症状的患者中,SC对复发具有良好的预测价值(AUROC为0.764,IC95为0.68-0.88),灵敏度为72%,特异性为77%,临界值为4.45µg/ml。SC和FC水平之间存在微弱但显著的相关性(r = 0.35, P = 0.001)。c反应蛋白、FC和SC联合评分对IBD的诊断没有效果。结论与有或无临床症状的内窥镜缓解患者相比,临床复发患者的sc明显升高。SC可以区分活动性IBD或IBS患者,但不能预测复发。
{"title":"Serum calprotectin is useful to confirm inflammatory bowel disease activity but not to predict relapse","authors":"Pauline Veyrard , Xavier Roblin , Céline Pansart , Ren Mao , Stéphane Nancey , Martin Killian , Louis Waeckel , Anne-Emmanuelle Berger , Nicolas Williet , Laetitia Bastide , Mathilde Barrau , Quentin Tournier , Stéphane Paul","doi":"10.1016/j.clicom.2022.02.001","DOIUrl":"10.1016/j.clicom.2022.02.001","url":null,"abstract":"<div><h3>Aim</h3><p>Serum calprotectin (SC), a novel biomarker of inflammatory bowel diseases (IBD), has been recently investigated with conflicting results. The purpose of this study was to assess the ability of SC to predict relapse in IBD patients treated by biologic therapies, and to evaluate the correlation between SC, clinical and endoscopic relapse and other biomarkers as fecal calprotectin (FC) and C-reactive protein (CRP).</p></div><div><h3>Methods</h3><p>All consecutive IBD patients in deep remission (clinical, endoscopic or imaging remission) were followed 12 months in this prospective study. Blood and stool samples were collected for SC, serum CRP and FC. SC was measured the day of inclusion (baseline, D0), 3 months (M3) and at 6 months (M6) or during the study period for clinical relapse. Relapse was defined as clinical, biomarkers, or endoscopic/imaging activities. Evolution of SC was quantified before relapse to analyze a predicting value of loss of response (LOR). SC for patients with active IBD and those with symptoms without inflammation were also compared.</p></div><div><h3>Results</h3><p>Among the 119 patients included, 54 (46.4%) patients experienced a disease relapse during follow-up. Median SC levels did not increase in patients with clinical relapse (3.15 µg/ml at baseline, 3.38 µg/ml at M3, 3.33 µg/ml at M6 and 3.99 µg/ml in case of relapse (<em>p</em> = 0.63)). SC were compared during relapse in patients with endoscopic remission but clinical symptoms defined as secondary Irritable Bowel Syndrome (IBS). SC levels were higher in active IBD and similar between the groups of patients with IBS or deep remission (3.05 µg/ml IBS <em>vs</em> 2.99 µg/ml remission <em>vs</em> 5.1 µg/ml for clinical relapse, <em>p</em> = 0.04). In patients with clinical symptoms, SC presents a good predictive value for relapse (AUROC 0.764, IC95: 0.68–0.88), with a sensitivity of 72%, a specificity of 77%, using a cut-off value of 4.45 µg/ml. A weak, but significant correlation was found between SC and FC levels (<em>r</em> = 0.35, <em>P</em> = 0.001). A combined score with CRP, FC and SC is not efficient to improve IBD diagnostic.</p></div><div><h3>Conclusion</h3><p>SC was significantly higher in patients with clinical relapse compared to those with endoscopic remission with or without clinical symptoms. SC allow to discriminate patients with active IBD or with IBS but failed to predict relapse.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S277261342200004X/pdfft?md5=39f3b7c1d3b877f7b7bb8e0e99a22f90&pid=1-s2.0-S277261342200004X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90691289","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2021.12.002
Zafran Khan , Ubaid Ahmad , Daniya Ualiyeva , Obed Boadi Amissah , Asaf Khan , Zohaib Noor , Nasib Zaman
SARS-CoV-2 causes Coronavirus Disease 2019 (COVID-19), an infectious condition that can present none or one or more of these symptoms: fever, cough, headache, sore throat, loss of taste and smell, aches, fatigue and musculoskeletal pain. For the prevention of COVID-19, there are vaccines available including those developed by Pfizer, Moderna, Sinovac, Janssen, and AstraZeneca. Recent evidence has shown that some COVID-19-vaccinated individuals can occasionally develop as a potential side effect Guillain-Barre syndrome (GBS), a severe neurological autoimmune condition in which the immune response against the peripheral nerve system (PNS) can result in significant morbidity. GBS had been linked previously to several viral or bacterial infections, and the finding of GBS after vaccination with certain COVID-19, while rare, should alert medical practitioners for an early diagnosis and targeted treatment. Here we review five cases of GBS that developed in different countries after COVID-19 vaccination.
{"title":"Guillain-Barre syndrome: An autoimmune disorder post-COVID-19 vaccination?","authors":"Zafran Khan , Ubaid Ahmad , Daniya Ualiyeva , Obed Boadi Amissah , Asaf Khan , Zohaib Noor , Nasib Zaman","doi":"10.1016/j.clicom.2021.12.002","DOIUrl":"10.1016/j.clicom.2021.12.002","url":null,"abstract":"<div><p>SARS-CoV-2 causes Coronavirus Disease 2019 (COVID-19), an infectious condition that can present none or one or more of these symptoms: fever, cough, headache, sore throat, loss of taste and smell, aches, fatigue and musculoskeletal pain. For the prevention of COVID-19, there are vaccines available including those developed by Pfizer, Moderna, Sinovac, Janssen, and AstraZeneca. Recent evidence has shown that some COVID-19-vaccinated individuals can occasionally develop as a potential side effect Guillain-Barre syndrome (GBS), a severe neurological autoimmune condition in which the immune response against the peripheral nerve system (PNS) can result in significant morbidity. GBS had been linked previously to several viral or bacterial infections, and the finding of GBS after vaccination with certain COVID-19, while rare, should alert medical practitioners for an early diagnosis and targeted treatment. Here we review five cases of GBS that developed in different countries after COVID-19 vaccination.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613421000147/pdfft?md5=f005b5e6811cb9abe9678febb5aa3d6a&pid=1-s2.0-S2772613421000147-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77059282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.09.005
Merve Hafızoğlu , Arzu Okyar Bas , Ece Tavukçuoğlu , Zeynep Sahiner , Merve Güner Oytun , Sıla Ulutürk , Hamdullah Yanık , Burcu Balam Doğu , Mustafa Cankurtaran , Güneş Esendağlı , Filiz Akbıyık , Banu Çakır , Serhat Ünal , Meltem Gülhan Halil
Generating memory T cell responses besides humoral immune responses is essential when it comes to the efficacy of a vaccine. In this study, the presence of memory T cell responses after aluminum-adjuvanted inactivated whole-virion SARS-CoV-2 vaccine (CoronaVac) in seronegative and seropositive elderly individuals were examined. CD4+ and CD8+ memory T cell proliferation and IFN-γ production capacities were evaluated. Additionally, clinical frailty scale (CFS) and FRAIL scales of the individuals were scored. CD4+ memory T cell responses more prominent than CD8+ memory T cells. In seronegative individuals, 80% of them had memory CD4+ and IFN-γ, whereas 50% of them had memory CD4+ and all of them had IFN-γ responses. Additionally, 40% of seronegative patients and 50% of seropositive patients had memory CD8+ responses. To sum up, humoral immune responses are not associated with memory T cell responses, and in seronegative individuals, memory T cell responses can be detected.
{"title":"Memory T cell responses in seronegative older adults following SARS-CoV-2 vaccination","authors":"Merve Hafızoğlu , Arzu Okyar Bas , Ece Tavukçuoğlu , Zeynep Sahiner , Merve Güner Oytun , Sıla Ulutürk , Hamdullah Yanık , Burcu Balam Doğu , Mustafa Cankurtaran , Güneş Esendağlı , Filiz Akbıyık , Banu Çakır , Serhat Ünal , Meltem Gülhan Halil","doi":"10.1016/j.clicom.2022.09.005","DOIUrl":"10.1016/j.clicom.2022.09.005","url":null,"abstract":"<div><p>Generating memory T cell responses besides humoral immune responses is essential when it comes to the efficacy of a vaccine. In this study, the presence of memory T cell responses after aluminum-adjuvanted inactivated whole-virion SARS-CoV-2 vaccine (CoronaVac) in seronegative and seropositive elderly individuals were examined. CD4<sup>+</sup> and CD8<sup>+</sup> memory T cell proliferation and IFN-γ production capacities were evaluated. Additionally, clinical frailty scale (CFS) and FRAIL scales of the individuals were scored. CD4<sup>+</sup> memory T cell responses more prominent than CD8<sup>+</sup> memory T cells. In seronegative individuals, 80% of them had memory CD4<sup>+</sup> and IFN-γ, whereas 50% of them had memory CD4<sup>+</sup> and all of them had IFN-γ responses. Additionally, 40% of seronegative patients and 50% of seropositive patients had memory CD8<sup>+</sup> responses. To sum up, humoral immune responses are not associated with memory T cell responses, and in seronegative individuals, memory T cell responses can be detected.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000257/pdfft?md5=56e3391f5c411316b885707b9433f9ef&pid=1-s2.0-S2772613422000257-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73061309","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.01.003
Samantha Woolery , Laura Bauler , Patrick Jones , Roua Azmeh
Adalimumab is a fully-humanized monoclonal antibody against tumor necrosis factor alpha (TNF-α) that is used to suppress the immune response. Though rare, hypersensitivity reactions to adalimumab can occur. Desensitization protocols have been developed to safely administer many medications in patients with Type I and Type IV hypersensitivity reactions. This case describes a teenage patient with Crohn's Disease who developed a delayed pruritic urticarial rash hypersensitivity reaction to adalimumab. A novel desensitization protocol was developed based on available literature. Our protocol consisted of four weekly doses, with the first dose in seven steps, second dose in two steps, and two full doses at weekly intervals, before spacing to biweekly injections. This multipart protocol was necessary due to the long half-life of adalimumab, which also allows for maintenance of the desensitized state in between injections. This protocol successfully treated our patient's hypersensitivity to adalimumab, with no further hypersensitivity occurring over a two-year follow-up period.
{"title":"Successful desensitization in a patient with adalimumab hypersensitivity","authors":"Samantha Woolery , Laura Bauler , Patrick Jones , Roua Azmeh","doi":"10.1016/j.clicom.2022.01.003","DOIUrl":"10.1016/j.clicom.2022.01.003","url":null,"abstract":"<div><p>Adalimumab is a fully-humanized monoclonal antibody against tumor necrosis factor alpha (TNF-α) that is used to suppress the immune response. Though rare, hypersensitivity reactions to adalimumab can occur. Desensitization protocols have been developed to safely administer many medications in patients with Type I and Type IV hypersensitivity reactions. This case describes a teenage patient with Crohn's Disease who developed a delayed pruritic urticarial rash hypersensitivity reaction to adalimumab. A novel desensitization protocol was developed based on available literature. Our protocol consisted of four weekly doses, with the first dose in seven steps, second dose in two steps, and two full doses at weekly intervals, before spacing to biweekly injections. This multipart protocol was necessary due to the long half-life of adalimumab, which also allows for maintenance of the desensitized state in between injections. This protocol successfully treated our patient's hypersensitivity to adalimumab, with no further hypersensitivity occurring over a two-year follow-up period.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000038/pdfft?md5=0d967616fb19734c7b38e9c7c7bd729c&pid=1-s2.0-S2772613422000038-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79703554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-12-01DOI: 10.1016/j.clicom.2022.08.002
Abhimanyu Thakur , Lifan Liang , Deepjyoti Ghosh , Alma Cili , Kui Zhang
Vitiligo is an autoimmune disorder, which is characterized by the chronic loss of melanocytes and subsequent lack of melanin from the skin or hair or both. The resultant depigmentation state of the skin causing irregular white patches deteriorates the vitiligo patients’ quality of life by major stigmatizing psychological impact. Despite tremendous studies for the past several decades, there has been a dearth of precise theranostics for vitiligo, which necessitates to revisit the molecular changes in vitiligo. Following advent of omics research, the examination of circulating non-coding RNAs including miRNAs has been enormously utilized for potential vitiligo therapeutic targets, however the expression profile of miRNAs and their target genes have not been studied at exosomal level in correlation with the differentially expressed genes (DEG) at the tissue level in vitiligo patients.
Exosomes are tiny extracellular vesicles, with diameter approximately 30–200 nm, which is released from various cell types, and found in different biofluids including blood serum and plasma. Notably, the exosomes have been found to mimic the constituents of their parent cells, enabling it an excellent theranostics platform for diseased condition. Therefore, we investigated the plasma exosomal miRNA and identified the target genes associated with immune infiltration in vitiligo patients compared to healthy subjects. In this study, 65 DEGs have been analyzed by heatmap, among which 44 genes are up-regulated and 21 are down-regulated, which are associated with melanin- biosynthetic and metabolic process. CENPN and SLIRP were found to be substantially correlated with immune cells viz. CD8+ T cells, M1-macrophage. Among several target genes by the exosomal miRNAs, SLIRP has been found to be associated with miR-16-5p, whereas CENPN has been found to be associated with miR-6721-5p, and miR-486-5p. Conclusively, the exosomal miR-16-5p, miR-6721-5p, and miR-486-5p could be potential theranostics targets for vitiligo.
{"title":"Identification and functional analysis of exosomal miR-16-5p, miR-6721-5p, and miR-486-5p associated with immune infiltration for potential vitiligo theranostics","authors":"Abhimanyu Thakur , Lifan Liang , Deepjyoti Ghosh , Alma Cili , Kui Zhang","doi":"10.1016/j.clicom.2022.08.002","DOIUrl":"10.1016/j.clicom.2022.08.002","url":null,"abstract":"<div><p>Vitiligo is an autoimmune disorder, which is characterized by the chronic loss of melanocytes and subsequent lack of melanin from the skin or hair or both. The resultant depigmentation state of the skin causing irregular white patches deteriorates the vitiligo patients’ quality of life by major stigmatizing psychological impact. Despite tremendous studies for the past several decades, there has been a dearth of precise theranostics for vitiligo, which necessitates to revisit the molecular changes in vitiligo. Following advent of omics research, the examination of circulating non-coding RNAs including miRNAs has been enormously utilized for potential vitiligo therapeutic targets, however the expression profile of miRNAs and their target genes have not been studied at exosomal level in correlation with the differentially expressed genes (DEG) at the tissue level in vitiligo patients.</p><p>Exosomes are tiny extracellular vesicles, with diameter approximately 30–200 nm, which is released from various cell types, and found in different biofluids including blood serum and plasma. Notably, the exosomes have been found to mimic the constituents of their parent cells, enabling it an excellent theranostics platform for diseased condition. Therefore, we investigated the plasma exosomal miRNA and identified the target genes associated with immune infiltration in vitiligo patients compared to healthy subjects. In this study, 65 DEGs have been analyzed by heatmap, among which 44 genes are up-regulated and 21 are down-regulated, which are associated with melanin- biosynthetic and metabolic process. <em>CENPN</em> and <em>SLIRP</em> were found to be substantially correlated with immune cells viz. CD8+ T cells, M1-macrophage. Among several target genes by the exosomal miRNAs, <em>SLIRP</em> has been found to be associated with miR-16-5p, whereas <em>CENPN</em> has been found to be associated with miR-6721-5p, and miR-486-5p. Conclusively, the exosomal miR-16-5p, miR-6721-5p, and miR-486-5p could be potential theranostics targets for vitiligo.</p></div>","PeriodicalId":100269,"journal":{"name":"Clinical Immunology Communications","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772613422000191/pdfft?md5=908570beb6fc1b603458566056826708&pid=1-s2.0-S2772613422000191-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82179865","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}