primary immunodeficiency disorders

  • 文章类型: Journal Article
    先前的研究表明,大多数患有先天性免疫错误(IEI)的患者在两次剂量的mRNA-1273COVID-19疫苗后会产生尖峰(S)特异性IgG抗体和T细胞反应,但是对加强疫苗接种的反应知之甚少。我们研究了171名IEI患者在使用基于mRNA的COVID-19疫苗加强疫苗接种8周后的免疫反应。此外,我们在荷兰COVID-19疫苗接种活动开始一年后评估了这些患者的临床结局.
    这项研究被纳入了一项大型前瞻性多中心研究,该研究调查了IEI中基于COVID-19mRNA的疫苗的免疫原性(VACOPID研究)。在加强免疫接种8周后,从244名参与者身上采集血液样本。这些参与者包括171名IEI患者(X连锁无丙种球蛋白血症(XLA;N=11),联合免疫缺陷(CID;N=4),常见可变免疫缺陷(CVID;N=45),孤立或未定义的抗体缺陷(N=108)和吞噬细胞缺陷(N=3))和73个对照。SARS-CoV-2特异性IgG滴度,中和抗体,和T细胞反应进行评估。COVID-19疫苗接种计划开始一年后,334名研究参与者(239名IEI患者和95名对照)完成了一份问卷,以补充他们的临床数据,重点是SARS-CoV-2感染。
    加强疫苗接种后,在所有COVID-19初始IEI队列和对照组中,S特异性IgG滴度增加,与启动方案后6个月的滴度相比。对照组和IEI队列之间的倍数增加没有差异。与启动方案后6个月相比,加强疫苗接种后的所有队列中SARS-CoV-2特异性T细胞反应也同样增加。在研究期间,大多数SARS-CoV-2感染都发生在Omicron变体占主导地位的时期。这些感染的临床过程是轻微的,尽管与对照组相比,IEI患者出现更频繁的发热和呼吸困难,并且他们的症状持续时间更长。
    我们的研究表明,在大多数IEI患者中,基于mRNA的加强疫苗接种可诱导记忆B细胞和T细胞反应的强烈回忆。一年的临床随访表明,IEI患者的SARS-CoV-2感染较轻。鉴于我们的结果,我们支持针对表型较温和的IEI患者的新型变体特异性COVID-19加强疫苗的加强运动.
    UNASSIGNED: Previous studies have demonstrated that the majority of patients with an inborn error of immunity (IEI) develop a spike (S)-specific IgG antibody and T-cell response after two doses of the mRNA-1273 COVID-19 vaccine, but little is known about the response to a booster vaccination. We studied the immune responses 8 weeks after booster vaccination with mRNA-based COVID-19 vaccines in 171 IEI patients. Moreover, we evaluated the clinical outcomes in these patients one year after the start of the Dutch COVID-19 vaccination campaign.
    UNASSIGNED: This study was embedded in a large prospective multicenter study investigating the immunogenicity of COVID-19 mRNA-based vaccines in IEI (VACOPID study). Blood samples were taken from 244 participants 8 weeks after booster vaccination. These participants included 171 IEI patients (X-linked agammaglobulinemia (XLA;N=11), combined immunodeficiency (CID;N=4), common variable immunodeficiency (CVID;N=45), isolated or undefined antibody deficiencies (N=108) and phagocyte defects (N=3)) and 73 controls. SARS-CoV-2-specific IgG titers, neutralizing antibodies, and T-cell responses were evaluated. One year after the start of the COVID-19 vaccination program, 334 study participants (239 IEI patients and 95 controls) completed a questionnaire to supplement their clinical data focusing on SARS-CoV-2 infections.
    UNASSIGNED: After booster vaccination, S-specific IgG titers increased in all COVID-19 naive IEI cohorts and controls, when compared to titers at 6 months after the priming regimen. The fold-increases did not differ between controls and IEI cohorts. SARS-CoV-2-specific T-cell responses also increased equally in all cohorts after booster vaccination compared to 6 months after the priming regimen. Most SARS-CoV-2 infections during the study period occurred in the period when the Omicron variant had become dominant. The clinical course of these infections was mild, although IEI patients experienced more frequent fever and dyspnea compared to controls and their symptoms persisted longer.
    UNASSIGNED: Our study demonstrates that mRNA-based booster vaccination induces robust recall of memory B-cell and T-cell responses in most IEI patients. One-year clinical follow-up demonstrated that SARS-CoV-2 infections in IEI patients were mild. Given our results, we support booster campaigns with newer variant-specific COVID-19 booster vaccines to IEI patients with milder phenotypes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胸腺植入是最近FDA批准的一种治疗先天性甲状腺功能异常的疗法。接受胸腺植入的患者会发展出功能性但不完整的T细胞区室。我们的目标是开发一种小鼠模型,以研究先天性异常中的临床胸腺植入,并优化植入程序,以最大程度地提高T细胞教育和原始T细胞的扩增。
    使用Foxn1nu无胸腺小鼠作为受体,我们测试了MHC匹配和不匹配的供体胸腺,它们作为新鲜组织植入或培养以去除供体T细胞.我们首先将胸腺植入肾囊下,然后优化肌内植入。使用竞争性过继转移试验,我们调查了新开发的T细胞未能扩增到完整的T细胞区室的原因是由于内在缺陷还是由于外周参与MHC分子的缺陷.最后,我们测试了重组IL-7是否会促进由植入的胸腺培养的宿主初始T细胞的扩增。
    我们确定Foxn1nu无胸腺小鼠的胸腺植入物模拟了先天性无胸腺综合征患者临床胸腺植入物的许多方面。当我们植入培养器时,MHC错配供体胸腺进入Foxn1nu无胸腺小鼠,小鼠发育出有限的T细胞区室,其原始种群明显不发达,记忆样T细胞过多。新产生的T细胞主要受供体胸腺表达的MHC分子的教育,因此,有可能在外周循环中进行另一轮选择。使用竞争性过继转移试验,我们比较了在MHC匹配和不匹配环境中接受供体胸腺教育的T细胞与在典型胸腺生成期间接受教育的T细胞的扩增率。一旦进入循环,无论MHC单倍型,在供体胸腺上接受教育的T细胞经历了异常扩增,最初更强劲的增殖以及更多的细胞死亡,类似于IL-7独立的自发扩增。用重组白细胞介素(IL-7)治疗植入的小鼠促进稳态扩增,改善T细胞发育,扩大了T细胞受体库,并使初始T细胞隔室正常化。
    我们得出的结论是,将培养的胸腺植入到Foxn1nu无胸腺小鼠的肌肉中是研究胸腺植入先天性无性系和免疫缺陷的合适系统。T细胞受供体胸腺的教育,然而,幼稚的T细胞在扩增方面存在缺陷。IL-7极大地改善了胸腺植入后的T细胞发育,并可能为改善临床胸腺植入的结果提供新的策略。
    UNASSIGNED: Thymus implantation is a recently FDA-approved therapy for congenital athymia. Patients receiving thymus implantation develop a functional but incomplete T cell compartment. Our objective was to develop a mouse model to study clinical thymus implantation in congenital athymia and to optimise implantation procedures to maximise T cell education and expansion of naïve T cells.
    UNASSIGNED: Using Foxn1 nu athymic mice as recipients, we tested MHC-matched and -mismatched donor thymi that were implanted as fresh tissue or cultured to remove donor T cells. We first implanted thymus under the kidney capsule and then optimised intramuscular implantation. Using competitive adoptive transfer assays, we investigated whether the failure of newly developed T cells to expand into a complete T cell compartment was because of intrinsic deficits or whether there were deficits in engaging MHC molecules in the periphery. Finally, we tested whether recombinant IL-7 would promote the expansion of host naïve T cells educated by the implanted thymus.
    UNASSIGNED: We determined that thymus implants in Foxn1 nu athymic mice mimic many aspects of clinical thymus implants in patients with congenital athymia. When we implanted cultured, MHC-mismatched donor thymus into Foxn1 nu athymic mice, mice developed a limited T cell compartment with notably underdeveloped naïve populations and overrepresented memory-like T cells. Newly generated T cells were predominantly educated by MHC molecules expressed by the donor thymus, thus potentially undergoing another round of selection once in the peripheral circulation. Using competitive adoptive transfer assays, we compared expansion rates of T cells educated on donor thymus versus T cells educated during typical thymopoiesis in MHC-matched and -mismatched environments. Once in the circulation, regardless of the MHC haplotypes, T cells educated on a donor thymus underwent abnormal expansion with initially more robust proliferation coupled with greater cell death, resembling IL-7 independent spontaneous expansion. Treating implanted mice with recombinant interleukin (IL-7) promoted homeostatic expansion that improved T cell development, expanded the T cell receptor repertoire, and normalised the naïve T cell compartment.
    UNASSIGNED: We conclude that implanting cultured thymus into the muscle of Foxn1 nu athymic mice is an appropriate system to study thymus implantation for congenital athymia and immunodeficiencies. T cells are educated by the donor thymus, yet naïve T cells have deficits in expansion. IL-7 greatly improves T cell development after thymus implantation and may offer a novel strategy to improve outcomes of clinical thymus implantation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Multicenter Study
    目标:先天性免疫错误(IEI)患者患严重冠状病毒病-2019(COVID-19)的风险增加。因此,有效的长期预防COVID-19对这些患者非常重要,但是对初次接种疫苗后免疫反应的衰减知之甚少。我们研究了473例IEI患者的两种mRNA-1273COVID-19疫苗接种后6个月的免疫反应,随后研究了50例常见可变免疫缺陷(CVID)患者对第三种mRNACOVID-19疫苗的反应。
    方法:在一项前瞻性多中心研究中,473例IEI患者(包括X连锁无丙种球蛋白血症(XLA)(N=18),联合免疫缺陷(CID)(N=22),CVID(N=203),孤立或未定义的抗体缺陷(N=204),和吞噬细胞缺陷(N=16)),179名对照纳入研究,并在两剂mRNA-1273COVID-19疫苗接种后随访6个月。此外,我们收集了50例CVID患者的样本,这些患者在通过国家疫苗接种计划进行初次疫苗接种后6个月接受了第三次疫苗接种.SARS-CoV-2特异性IgG滴度,中和抗体,评估T细胞反应。
    结果:接种后6个月,IEI患者和健康对照的几何平均抗体滴度(GMT)均下降,与疫苗接种后28天的GMT相比。这种下降的轨迹在对照组和大多数IEI队列之间没有差异;然而,CID中的抗体滴度,CVID,与对照组相比,分离的抗体缺乏症患者更经常下降到低于反应者的临界值。在接种后6个月,在77%的对照和68%的IEI患者中仍可检测到特异性T细胞应答。第三种mRNA疫苗在30名CVID患者中仅有两名在两种mRNA疫苗后没有血清转化的患者中产生抗体应答。
    结论:在接种mRNA-1273COVID-19后6个月,与健康对照组相比,IEI患者的IgG滴度和T细胞反应下降相似。第三种mRNACOVID-19疫苗对先前无反应的CVID患者的有益益处有限,这表明这些脆弱的患者需要其他保护策略。
    Patients with inborn errors of immunity (IEI) are at increased risk of severe coronavirus disease-2019 (COVID-19). Effective long-term protection against COVID-19 is therefore of great importance in these patients, but little is known about the decay of the immune response after primary vaccination. We studied the immune responses 6 months after two mRNA-1273 COVID-19 vaccines in 473 IEI patients and subsequently the response to a third mRNA COVID-19 vaccine in 50 patients with common variable immunodeficiency (CVID).
    In a prospective multicenter study, 473 IEI patients (including X-linked agammaglobulinemia (XLA) (N = 18), combined immunodeficiency (CID) (N = 22), CVID (N = 203), isolated or undefined antibody deficiencies (N = 204), and phagocyte defects (N = 16)), and 179 controls were included and followed up to 6 months after two doses of the mRNA-1273 COVID-19 vaccine. Additionally, samples were collected from 50 CVID patients who received a third vaccine 6 months after primary vaccination through the national vaccination program. SARS-CoV-2-specific IgG titers, neutralizing antibodies, and T cell responses were assessed.
    At 6 months after vaccination, the geometric mean antibody titers (GMT) declined in both IEI patients and healthy controls, when compared to GMT 28 days after vaccination. The trajectory of this decline did not differ between controls and most IEI cohorts; however, antibody titers in CID, CVID, and isolated antibody deficiency patients more often dropped to below the responder cut-off compared to controls. Specific T cell responses were still detectable in 77% of controls and 68% of IEI patients at 6 months post vaccination. A third mRNA vaccine resulted in an antibody response in only two out of 30 CVID patients that did not seroconvert after two mRNA vaccines.
    A similar decline in IgG titers and T cell responses was observed in patients with IEI when compared to healthy controls 6 months after mRNA-1273 COVID-19 vaccination. The limited beneficial benefit of a third mRNA COVID-19 vaccine in previous non-responder CVID patients implicates that other protective strategies are needed for these vulnerable patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    未经证实:自身免疫性多内分泌病-念珠菌病-外胚层营养不良(APECED)和与肌腱挛缩相关的多毛皮病,肌病,和肺纤维化(POIKTMP)是罕见的遗传综合征,由AIRE的双等位基因致病性变异和FAM111B的杂合致病性变异引起,分别。APECED和POIKTMP的临床诊断依赖于定义相应综合征的两种或多种特征性疾病表现的发展。我们讨论了共同和独特的临床,射线照相,和APECED和POIKTMP之间的组织学特征,并描述了他对硫唑嘌呤对POIKTMP相关肝炎的治疗反应,肌炎,和肺炎。
    UASSIGNED:通过知情同意和纳入IRB批准的方案(NCT01386437,NCT03206099),患者在NIH临床中心进行了全面的临床评估,同时进行了外显子组测序,拷贝数变异分析,自身抗体调查,外周血免疫分型,和唾液细胞因子分析。
    UNASSIGNED:我们报告了一名9岁男孩的报告和评估,该男孩被转诊到美国国立卫生研究院临床中心,具有APECED样临床表型,包括经典的APECEDCMC和甲状旁腺功能减退。他被发现符合以真皮病为特征的POIKTMP的临床诊断标准,肌腱挛缩,肌病,和肺炎,和外显子组测序显示FAM111B中的从头c.1292T>C杂合致病变异,但AIRE中没有有害的单核苷酸变异或拷贝数变异。
    未经评估:本报告扩展了现有的遗传,临床,自身抗体,免疫学,以及关于POIKTMP的治疗反应信息。
    Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) and poikiloderma in association with tendon contractures, myopathy, and pulmonary fibrosis (POIKTMP) are rare inherited syndromes resulting from biallelic pathogenic variants in AIRE and heterozygous pathogenic variants in FAM111B, respectively. The clinical diagnosis of APECED and POIKTMP rely on the development of two or more characteristic disease manifestations that define the corresponding syndromes. We discuss the shared and distinct clinical, radiographic, and histological features between APECED and POIKTMP presented in our patient case and describe his treatment response to azathioprine for POIKTMP-associated hepatitis, myositis, and pneumonitis.
    Through informed consent and enrollment onto IRB-approved protocols (NCT01386437, NCT03206099) the patient underwent a comprehensive clinical evaluation at the NIH Clinical Center alongside exome sequencing, copy number variation analysis, autoantibody surveys, peripheral blood immunophenotyping, and salivary cytokine analyses.
    We report the presentation and evaluation of a 9-year-old boy who was referred to the NIH Clinical Center with an APECED-like clinical phenotype that included the classic APECED dyad of CMC and hypoparathyroidism. He was found to meet clinical diagnostic criteria for POIKTMP featuring poikiloderma, tendon contractures, myopathy, and pneumonitis, and exome sequencing revealed a de novo c.1292T>C heterozygous pathogenic variant in FAM111B but no deleterious single nucleotide variants or copy number variants in AIRE.
    This report expands upon the available genetic, clinical, autoantibody, immunological, and treatment response information on POIKTMP.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    原发性免疫缺陷疾病包括一组罕见的主要是由先天性免疫错误引起的单基因疾病。大多数可以通过Sanger测序或下一代测序来鉴定。一些病症由导致拷贝数变异(CNV)的大插入或缺失引起。Sanger测序可能无法识别这些突变。在这里,我们提出了液滴数字PCR作为一种替代的经济有效的诊断方法来识别这些基因中的CNV。提供了NFKB1,SERPING1和SH2D1A大缺失患者的数据。
    Primary immunodeficiency disorders comprise a rare group of mostly monogenic disorders caused by inborn errors of immunity. The majority can be identified by either Sanger sequencing or next generation sequencing. Some disorders result from large insertions or deletions leading to copy number variations (CNVs). Sanger sequencing may not identify these mutations. Here we present droplet digital PCR as an alternative cost-effective diagnostic method to identify CNV in these genes. The data from patients with large deletions of NFKB1, SERPING1, and SH2D1A are presented.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Controlled Clinical Trial
    先天性免疫错误(IEI)患者患严重冠状病毒病-2019(COVID-19)的风险增加。因此,针对COVID-19的有效疫苗接种在这一群体中非常重要,但对COVID-19疫苗在这些患者中的免疫原性知之甚少。
    我们试图研究成年IEI患者在mRNA-1273COVID-19疫苗接种后的体液和细胞免疫反应。
    在未来,控制,多中心研究,505例IEI患者(常见可变免疫缺陷[CVID],孤立或未定义的抗体缺乏,X-连锁丙种球蛋白血症,联合B细胞和T细胞免疫缺陷,吞噬细胞缺陷)和192个对照被包括在内。所有参与者都接受了2剂mRNA-1273COVID-19疫苗。严重急性呼吸综合征冠状病毒-2特异性结合抗体的水平,中和抗体,在基线时评估T细胞反应,第一次接种疫苗后28天,第二次接种疫苗后28天。
    临床轻度抗体缺乏和吞噬细胞缺陷患者的血清转换率与健康对照组相似,但是IEI更严重的患者的血清转换率,如CVID和B细胞和T细胞联合免疫缺陷,较低。结合抗体滴度与中和抗体的存在密切相关。T细胞反应与所有IEI队列中的对照组相当,除CVID患者外。CVID患者的非感染性并发症的存在和免疫抑制药物的使用与抗体反应呈负相关。
    用mRNA-1273接种COVID-19在轻度抗体缺乏和吞噬细胞缺陷以及大多数患有B细胞和T细胞免疫缺陷和CVID的患者中具有免疫原性。在X连锁无丙种球蛋白血症患者和具有非感染性并发症的CVID患者中检测到最低反应。评估这些脆弱患者群体中免疫反应的寿命将指导进一步接种疫苗的决策。
    Patients with inborn errors of immunity (IEI) are at increased risk of severe coronavirus disease-2019 (COVID-19). Effective vaccination against COVID-19 is therefore of great importance in this group, but little is known about the immunogenicity of COVID-19 vaccines in these patients.
    We sought to study humoral and cellular immune responses after mRNA-1273 COVID-19 vaccination in adult patients with IEI.
    In a prospective, controlled, multicenter study, 505 patients with IEI (common variable immunodeficiency [CVID], isolated or undefined antibody deficiencies, X-linked agammaglobulinemia, combined B- and T-cell immunodeficiency, phagocyte defects) and 192 controls were included. All participants received 2 doses of the mRNA-1273 COVID-19 vaccine. Levels of severe acute respiratory syndrome coronavirus-2-specific binding antibodies, neutralizing antibodies, and T-cell responses were assessed at baseline, 28 days after first vaccination, and 28 days after second vaccination.
    Seroconversion rates in patients with clinically mild antibody deficiencies and phagocyte defects were similar to those in healthy controls, but seroconversion rates in patients with more severe IEI, such as CVID and combined B- and T-cell immunodeficiency, were lower. Binding antibody titers correlated well to the presence of neutralizing antibodies. T-cell responses were comparable to those in controls in all IEI cohorts, with the exception of patients with CVID. The presence of noninfectious complications and the use of immunosuppressive drugs in patients with CVID were negatively correlated with the antibody response.
    COVID-19 vaccination with mRNA-1273 was immunogenic in mild antibody deficiencies and phagocyte defects and in most patients with combined B- and T-cell immunodeficiency and CVID. Lowest response was detected in patients with X-linked agammaglobulinemia and in patients with CVID with noninfectious complications. The assessment of longevity of immune responses in these vulnerable patient groups will guide decision making for additional vaccinations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: Severe combined immunodeficiency (SCID) is a group of relatively rare primary immunodeficiency disorders (PIDs), characterized by disturbed development of T cells and B cells, caused by several genetic mutations that bring on different clinical presentations. SCID may be inherited as an autosomal recessive or an X-linked genetic trait.
    METHODS: A 6-year-old male presented with a history of food allergy, productive coughs, and recurrent purulent rhinitis, poor weight gain and hypothyroidism. The total count of CD4+ T lymphocytes, along with their naïve and central memory subpopulations, as well as central memory CD8+ T cells were decreased in flow cytometry. A nucleotide substitution in exon one of interleukin 2 receptor gamma chain (IL-2RG) gene (c.115 G>A, p.D39N, ChrX: 70,331,275) was reported, based on which the diagnosis of X-liked SCID was confirmed. Antiviral and antibiotic prophylaxis, along with monthly IVIG (intravenous immunoglobulin) was started and the patient was subsequently referred for hematopoietic stem cell transplantation.
    CONCLUSIONS: PIDs should be considered as the differential diagnosis in any patient with unexplained and bizarre symptoms associated with recurrent infections, allergic and autoimmune manifestations. Clinicians should also bear X-SCID in mind in case of approach to any patient with poor weight gain, unusual allergic or endocrine manifestations, even in the case of a normal or increased level of serum immunoglobulins or T and B cells numbers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    Severe Combined Immunodeficiency (SCID) involves the lymphocyte lineage and mimics Human Immunodeficiency Virus (HIV) disease common in our region, making it difficult to diagnose and manage effectively. SCID in East Africa stands underdiagnosed because of lack of awareness and diagnostic resources. A case series of three SCID patients admitted to a Tertiary Paediatric Centre in Kenya between 2016 and 2019. The clinical presentations, laboratory findings, management and outcome for each were studied. Three cases were diagnosed between the ages of 4 to 15 months. Two of them were male and one was a female. All had a history of previous sibling death. There was no parental consanguinity. All presented with pneumonia. One of them had vaccine acquired Rotavirus infection and a persistent generalised maculopapular rash. The T, B cell profile was T- B- in two and T- B+ in one case and the immunoglobulins were reduced in all. All the cases were fatal. Thus, Primary immunodeficiency disorders are prevalent in East Africa. A proper clinical history, examination and laboratory tests like a haemogram, peripheral blood film can aid to suspect and diagnose SCID even with limited resources.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    2020年12月中旬,以色列开始在全国范围内开展针对2019年冠状病毒病(COVID-19)的大规模疫苗接种运动。在最初的几周里,医务人员,老年公民,慢性病患者优先。因此,我们鼓励患有原发性和继发性免疫缺陷的患者接种疫苗.尽管基于RNA的COVID-19疫苗的功效已在普通人群中得到证实,对先天性免疫错误(IEI)患者的疗效和安全性知之甚少.
    我们的目的是评估一组IEI患者对COVID-19疫苗的体液和细胞免疫反应。
    共纳入26名成人患者,在第二剂Pfizer-BioNTechCOVID-19疫苗接种后2周,从中收集血浆和外周血单核细胞。通过测试抗SARS-CoV-2刺突(S)受体结合域和抗核衣壳抗体滴度并通过抑制受体结合域-血管紧张素转换酶2结合来评估中和能力来评估体液反应。通过使用ELISpot评估细胞免疫反应,评估响应于合并的SARS-CoV-2S或M肽的IL-2和IFN-γ分泌。
    我们的队列包括18名主要为抗体缺乏的患者,2合并免疫缺陷,3与免疫失调,和3与其他基因定义的诊断。其中22人正在接受免疫球蛋白替代疗法。在26名患者中,18开发了特异性抗体反应,和19显示S肽特异性T细胞应答。没有患者报告显著的不良事件。
    为IEI患者接种疫苗是安全的,大多数患者能够产生疫苗特异性抗体反应,S蛋白特异性细胞反应,或者两者兼而有之。
    In mid-December 2020, Israel started a nationwide mass vaccination campaign against coronavirus disease 2019 (COVID-19). In the first few weeks, medical personnel, elderly citizens, and patients with chronic diseases were prioritized. As such, patients with primary and secondary immunodeficiencies were encouraged to receive the vaccine. Although the efficacy of RNA-based COVID-19 vaccines has been demonstrated in the general population, little is known about their efficacy and safety in patients with inborn errors of immunity (IEI).
    Our aim was to evaluate the humoral and cellular immune response to COVID-19 vaccine in a cohort of patients with IEI.
    A total of 26 adult patients were enrolled, and plasma and peripheral blood mononuclear cells were collected from them 2 weeks following the second dose of Pfizer-BioNTech COVID-19 vaccine. Humoral response was evaluated by testing anti-SARS-CoV-2 spike (S) receptor-binding domain and antinucleocapsid antibody titers and evaluating neutralizing ability by inhibition of receptor-binding domain-angiotensin-converting enzyme 2 binding. Cellular immune response was evaluated by using ELISpot, estimating IL-2 and IFN-γ secretion in response to pooled SARS-CoV-2 S- or M-peptides.
    Our cohort included 18 patients with a predominantly antibody deficiency, 2 with combined immunodeficiency, 3 with immune dysregulation, and 3 with other genetically defined diagnoses. Twenty-two of them were receiving immunoglobulin replacement therapy. Of the 26 patients, 18 developed specific antibody response, and 19 showed S-peptide-specific T-cell response. None of the patients reported significant adverse events.
    Vaccinating patients with IEI is safe, and most patients were able to develop vaccine-specific antibody response, S-protein-specific cellular response, or both.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    大多数儿童免疫失调的患者,多内分泌病,肠病没有基因诊断。这些患者可能接受经验性免疫抑制治疗,结果差异很大。
    我们试图确定免疫失调患者疾病的遗传基础,多内分泌病,肠病,X连锁样(IPEX样)疾病,但FOXP3无突变;然后评估基因诊断对临床治疗的影响。
    使用一组462个与先天免疫错误有关的基因对基因组DNA进行测序。候选突变的特征是基因组,转录,和(对于某些)蛋白质分析。
    在123例FOXP3阴性IPEX样疾病患者中,48(39%)在以下27个基因中的1个中携带了破坏性的种系突变:AIRE,BACH2,BCL11B,CARD11,CARD14,CTLA4,IRF2BP2,ITCH,JAK1,KMT2D,LRBA,MYO5B,NFKB1,NLRC4,POLA1,POMP,RAG1,SH2D1A,SKIV2L,STAT1,STAT3,TNFAIP3,TNFRSF6/FAS,TNRSF13B/TACI、TOM1、TTC37和XIAP。这些基因中的许多基因以前与IPEX样诊断无关。对于48例基因诊断患者中的42例,知道关键基因可能会改变治疗管理,包括靶向治疗和支持或反对造血细胞移植的建议。
    现在被捆绑为“IPEX样”疾病的许多儿童疾病都是由个别罕见的疾病引起的,免疫调节基因的严重突变。这些病症的大多数遗传诊断产生临床上可操作的发现。如果旧技术未能提供诊断,则障碍是缺乏测试或缺乏重复测试。
    Most patients with childhood-onset immune dysregulation, polyendocrinopathy, and enteropathy have no genetic diagnosis for their illness. These patients may undergo empirical immunosuppressive treatment with highly variable outcomes.
    We sought to determine the genetic basis of disease in patients referred with Immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like (IPEX-like) disease, but with no mutation in FOXP3; then to assess consequences of genetic diagnoses for clinical management.
    Genomic DNA was sequenced using a panel of 462 genes implicated in inborn errors of immunity. Candidate mutations were characterized by genomic, transcriptional, and (for some) protein analysis.
    Of 123 patients with FOXP3-negative IPEX-like disease, 48 (39%) carried damaging germline mutations in 1 of the following 27 genes: AIRE, BACH2, BCL11B, CARD11, CARD14, CTLA4, IRF2BP2, ITCH, JAK1, KMT2D, LRBA, MYO5B, NFKB1, NLRC4, POLA1, POMP, RAG1, SH2D1A, SKIV2L, STAT1, STAT3, TNFAIP3, TNFRSF6/FAS, TNRSF13B/TACI, TOM1, TTC37, and XIAP. Many of these genes had not been previously associated with an IPEX-like diagnosis. For 42 of the 48 patients with genetic diagnoses, knowing the critical gene could have altered therapeutic management, including recommendations for targeted treatments and for or against hematopoietic cell transplantation.
    Many childhood disorders now bundled as \"IPEX-like\" disease are caused by individually rare, severe mutations in immune regulation genes. Most genetic diagnoses of these conditions yield clinically actionable findings. Barriers are lack of testing or lack of repeat testing if older technologies failed to provide a diagnosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号