Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
J Clin Immunol ; 43(5): 965-978, 2023 07.
Article in English | MEDLINE | ID: mdl-36843153

ABSTRACT

BACKGR OUND: T-cell receptor excision circle (TREC)-based newborn screening (NBS) for severe combined immunodeficiencies (SCID) was introduced in Germany in August 2019. METHODS: Children with abnormal TREC-NBS were referred to a newly established network of Combined Immunodeficiency (CID) Clinics and Centers. The Working Group for Pediatric Immunology (API) and German Society for Newborn Screening (DGNS) performed 6-monthly surveys to assess the TREC-NBS process after 2.5 years. RESULTS: Among 1.9 million screened newborns, 88 patients with congenital T-cell lymphocytopenia were identified (25 SCID, 17 leaky SCID/Omenn syndrome (OS)/idiopathic T-cell lymphocytopenia, and 46 syndromic disorders). A genetic diagnosis was established in 88%. Twenty-six patients underwent hematopoietic stem cell transplantation (HSCT), 23/26 within 4 months of life. Of these, 25/26 (96%) were alive at last follow-up. Two patients presented with in utero onset OS and died after birth. Five patients with syndromic disorders underwent thymus transplantation. Eight syndromic patients deceased, all from non-immunological complications. TREC-NBS missed one patient, who later presented clinically, and one tracking failure occurred after an inconclusive screening result. CONCLUSION: The German TREC-NBS represents the largest European SCID screening at this point. The incidence of SCID/leaky SCID/OS in Germany is approximately 1:54,000, very similar to previous observations from North American and European regions and countries where TREC-NBS was implemented. The newly founded API-CID network facilitates tracking and treatment of identified patients. Short-term HSCT outcome was excellent, but NBS and transplant registries will remain essential to evaluate the long-term outcome and to compare results across the rising numbers of TREC-NBS programs across Europe.


Subject(s)
Lymphopenia , Severe Combined Immunodeficiency , Child , Infant, Newborn , Humans , Neonatal Screening/methods , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/therapy , Prospective Studies , Lymphopenia/diagnosis , DNA , Germany/epidemiology , Receptors, Antigen, T-Cell/genetics
2.
Infection ; 51(5): 1357-1367, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36787015

ABSTRACT

PURPOSE: SARS-CoV-2 infections cause COVID-19 and have a wide spectrum of morbidity. Severe disease courses among children are rare. To date, data on the variability of morbidity in relation to variant of concern (VOC) in children has been sparse and inconclusive. We compare the clinical severity of SARS-CoV-2 infection among children and adolescents in Germany during the Wildtype and Alpha combined, Delta and Omicron phases of the COVID-19 pandemic. METHODS: Comparing risk of COVID-19-related hospitalization, intensive care unit (ICU) admission and death due to COVID-19 in children and adolescents, we used: (1) a multi-center seroprevalence study (SARS-CoV-2-KIDS study); (2) a nationwide registry of pediatric patients hospitalized with SARS-CoV-2 infections; and (3) compulsory national reporting for RT-PCR-confirmed SARS-CoV-2 infections in Germany. RESULTS: During the Delta predominant phase, risk of COVID-19-related hospitalization among all SARS-CoV-2 seropositive children was 3.35, ICU admission 1.19 and fatality 0.09 per 10,000; hence about halved for hospitalization and ICU admission and unchanged for deaths as compared to the Wildtype- and Alpha-dominant period. The relative risk for COVID-19-related hospitalization and ICU admission compared to the alpha period decreased during Delta [0.60 (95% CI 0.54; 0.67) and 0.51 (95% CI 0.42; 0.61)] and Omicron [0.27 (95% CI 0.24; 0.30) and 0.06 (95% CI 0.05; 0.08)] period except for the < 5-year-olds. The rate of case fatalities decreased slightly during Delta, and substantially during Omicron phase. CONCLUSION: Morbidity caused by SARS-CoV-2 infections among children and adolescents in Germany decreased over the course of the COVID-19 pandemic, as different VOCs) emerged.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Adolescent , Child , Child, Preschool , COVID-19/epidemiology , Risk , Pandemics , Seroepidemiologic Studies , Hospitalization , Germany/epidemiology , Intensive Care Units
3.
Clin Immunol ; 245: 109143, 2022 12.
Article in English | MEDLINE | ID: mdl-36191738

ABSTRACT

BACKGROUND: New therapeutic strategies for juvenile idiopathic arthritis (JIA) have evolved within the past ten years, and as a result, an update of the 2011 recommendations of the German management guidelines was initiated. METHODS: A systemic literature review was performed, overarching principles were proposed and pre-selected via an online survey followed by two multidisciplinary consensus conferences. Pharmacological and non-pharmacological treatments were discussed, statements were proposed and ultimately agreed upon by nominal group technique (NGT). RESULTS: 12 overarching therapeutic principles, as well as 9 recommendations on pharmacological and 5 on non-pharmacological treatments for JIA were agreed upon. CONCLUSION: This report summarizes the recent update of the interdisciplinary, consensus-based German guidelines on the management of JIA. The multi- and interdisciplinary participation of all caregivers was central for this patient-focused update. With these guidelines, physicians can choose an evidence-based approach, which allows better tailored treatment in this vulnerable cohort of children and adolescents.


Subject(s)
Arthritis, Juvenile , Adolescent , Child , Humans , Arthritis, Juvenile/drug therapy , Consensus , Developmental Disabilities
4.
J Clin Immunol ; 41(8): 1878-1892, 2021 11.
Article in English | MEDLINE | ID: mdl-34477998

ABSTRACT

Patients with ataxia-telangiectasia (A-T) suffer from progressive cerebellar ataxia, immunodeficiency, respiratory failure, and cancer susceptibility. From a clinical point of view, A-T patients with IgA deficiency show more symptoms and may have a poorer prognosis. In this study, we analyzed mortality and immunity data of 659 A-T patients with regard to IgA deficiency collected from the European Society for Immunodeficiencies (ESID) registry and from 66 patients with classical A-T who attended at the Frankfurt Goethe-University between 2012 and 2018. We studied peripheral B- and T-cell subsets and T-cell repertoire of the Frankfurt cohort and survival rates of all A-T patients in the ESID registry. Patients with A-T have significant alterations in their lymphocyte phenotypes. All subsets (CD3, CD4, CD8, CD19, CD4/CD45RA, and CD8/CD45RA) were significantly diminished compared to standard values. Patients with IgA deficiency (n = 35) had significantly lower lymphocyte counts compared to A-T patients without IgA deficiency (n = 31) due to a further decrease of naïve CD4 T-cells, central memory CD4 cells, and regulatory T-cells. Although both patient groups showed affected TCR-ß repertoires compared to controls, no differences could be detected between patients with and without IgA deficiency. Overall survival of patients with IgA deficiency was significantly diminished. For the first time, our data show that patients with IgA deficiency have significantly lower lymphocyte counts and subsets, which are accompanied with reduced survival, compared to A-T patients without IgA deficiency. IgA, a simple surrogate marker, is indicating the poorest prognosis for classical A-T patients. Both non-interventional clinical trials were registered at clinicaltrials.gov 2012 (Susceptibility to infections in ataxia-telangiectasia; NCT02345135) and 2017 (Susceptibility to Infections, tumor risk and liver disease in patients with ataxia-telangiectasia; NCT03357978).


Subject(s)
Ataxia Telangiectasia/immunology , Ataxia Telangiectasia/mortality , B-Lymphocytes/immunology , IgA Deficiency/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Humans , IgA Deficiency/mortality , IgG Deficiency/immunology , IgG Deficiency/mortality , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant , Lymphocyte Count , Male , Middle Aged , Young Adult
5.
Eur J Immunol ; 50(10): 1432-1446, 2020 10.
Article in English | MEDLINE | ID: mdl-32845010

ABSTRACT

This evidence-based clinical guideline provides consensus-recommendations for the treatment and care of patients with primary antibody deficiencies (PADs). The guideline group comprised 20 clinical and scientific expert associations of the German, Swiss, and Austrian healthcare system and representatives of patients. Recommendations were based on results of a systematic literature search, data extraction, and evaluation of methodology and study quality in combination with the clinical expertise of the respective representatives. Consensus-based recommendations were determined via nominal group technique. PADs are the largest clinically relevant group of primary immunodeficiencies. Most patients with PADs present with increased susceptibility to infections, however immune dysregulation, autoimmunity, and cancer affect a significant number of patients and may precede infections. This guideline therefore covers interdisciplinary clinical and therapeutic aspects of infectious (e.g., antibiotic prophylaxis, management of bronchiectasis) and non-infectious manifestations (e.g., management of granulomatous disease, immune cytopenia). PADs are grouped into disease entities with definitive, probable, possible, or unlikely benefit of IgG-replacement therapy. Summary and consensus-recommendations are provided for treatment indication, dosing, routes of administration, and adverse events of IgG-replacement therapy. Special aspects of concomitant impaired T-cell function are highlighted as well as clinical data on selected monogenetic inborn errors of immunity formerly classified into PADs (APDS, CTLA-4-, and LRBA-deficiency).


Subject(s)
Immunoglobulins/therapeutic use , Primary Immunodeficiency Diseases/therapy , Austria , Autoimmunity , Consensus , Drug-Related Side Effects and Adverse Reactions , Evidence-Based Medicine , Germany , Humans , Interdisciplinary Communication , Practice Guidelines as Topic , Primary Immunodeficiency Diseases/immunology , Switzerland
6.
Eur J Immunol ; 50(7): 1078-1080, 2020 07.
Article in English | MEDLINE | ID: mdl-32181500

ABSTRACT

AMPK (adenosine monophosphate-activated protein kinase) is phosphorylated (AMPK-P) in response to low energy through allosteric activation by Adenosine mono- or diphosphate (AMP/ADP). Folliculin (FLCN) and the FLCN-interacting proteins 1 and 2 (FNIP1, 2) modulate AMPK. FNIP1 deficiency patients have a AMPK-P gain of function phenotype with hypertrophic cardiomyopathy, Wolff-Parkinson-White pre-excitation syndrome, myopathy of skeletal muscles and combined immunodeficiency.


Subject(s)
Cardiomyopathies , Carrier Proteins , Genes, Recessive , Immunologic Deficiency Syndromes , Mutation , Pre-Excitation Syndromes , Cardiomyopathies/genetics , Cardiomyopathies/immunology , Cardiomyopathies/pathology , Carrier Proteins/genetics , Carrier Proteins/immunology , Female , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Male , Pre-Excitation Syndromes/genetics , Pre-Excitation Syndromes/immunology , Pre-Excitation Syndromes/pathology
7.
Clin Infect Dis ; 71(10): 2561-2569, 2020 12 17.
Article in English | MEDLINE | ID: mdl-31796965

ABSTRACT

BACKGROUND: In adults, anti-tumor necrosis factor-α (TNF-α) therapy is associated with progression of latent tuberculosis (TB) infection (LTBI) to TB disease, but pediatric data are limited. METHODS: Retrospective multicenter study within the Paediatric Tuberculosis Network European Trials Group, capturing patients <18 years who developed TB disease during anti-TNF-α therapy. RESULTS: Sixty-six tertiary healthcare institutions providing care for children with TB participated. Nineteen cases were identified: Crohn's disease (n = 8; 42%) and juvenile idiopathic arthritis (n = 6; 32%) were the commonest underlying conditions. Immune-based TB screening (tuberculin skin test and/or interferon-γ release assay) was performed in 15 patients before commencing anti-TNF-α therapy but only identified 1 LTBI case; 13 patients were already receiving immunosuppressants at the time of screening. The median interval between starting anti-TNF-α therapy and TB diagnosis was 13.1 (IQR, 7.1-20.3) months. All cases presented with severe disease, predominantly miliary TB (n = 14; 78%). One case was diagnosed postmortem. TB was microbiologically confirmed in 15 cases (79%). The median duration of anti-TB treatment was 50 (IQR, 46-66) weeks. Five of 15 (33%) cases who had completed TB treatment had long-term sequelae. CONCLUSIONS: LTBI screening is frequently false-negative in this patient population, likely due to immunosuppressants impairing test performance. Therefore, patients with immune-mediated diseases should be screened for LTBI at the point of diagnosis, before commencing immunosuppressive medication. Children on anti-TNF-α therapy are prone to severe TB disease and significant long-term morbidity. Those observations underscore the need for robust LTBI screening programs in this high-risk patient population, even in low-TB-prevalence settings.


Subject(s)
Latent Tuberculosis , Tuberculosis , Adolescent , Adult , Child , Humans , Interferon-gamma Release Tests , Latent Tuberculosis/diagnosis , Latent Tuberculosis/epidemiology , Necrosis , Retrospective Studies , Tuberculin Test , Tuberculosis/epidemiology , Tumor Necrosis Factor-alpha
8.
J Clin Immunol ; 40(5): 708-717, 2020 07.
Article in English | MEDLINE | ID: mdl-32458183

ABSTRACT

PURPOSE: Severe combined immunodeficiencies (SCID) are a heterogeneous group of fatal genetic disorders, in which the immune response is severely impaired. SCID can be cured if diagnosed early. We aim to determine the incidence of clinically defined SCID cases, acquire data of reported cases and evaluate their possible prediction by newborn screening, before introduction of a general screening program in Germany. METHODS: The German Surveillance Unit for rare Paediatric Diseases (ESPED) prospectively queried the number of incident SCID cases in all German paediatric hospitals in 2014 and 2015. Inclusion criteria were (1) opportunistic or severe infections or clinical features associated with SCID (failure to thrive, lacking thymus or lymphatic tissue, dysregulation of the immune system, graft versus host reaction caused by maternal T cells), (2) dysfunctional T cell immunity or proof of maternal T cells and (3) exclusion of a secondary immunodeficiency such as human immunodeficiency virus (HIV) infection. In a capture-recapture analysis, cases were matched with cases reported to the European Society for Immunodeficiencies (ESID). RESULTS: Fifty-eight patients were initially reported to ESPED, 24 reports could be confirmed as SCID, 21 patients were less than 1 year old at time of diagnosis. One SCID case was reported to ESID only. The estimated incidence of SCID in Germany is 1.6/100,000 (1:62,500) per year in children less than 1 year of age. Most patients reported were symptomatic and mortality in regard to reported outcome was high (29% (6/22)). The majority of incident SCID cases were considered to be probably detectable by newborn screening. CONCLUSIONS: SCID is a rare disease with significant mortality. Newborn screening may give the opportunity to improve the prognosis in a significant number of children with SCID.


Subject(s)
Severe Combined Immunodeficiency/epidemiology , Female , Germany/epidemiology , Humans , Incidence , Infant , Infant, Newborn , Male , Neonatal Screening , Phenotype , Severe Combined Immunodeficiency/mortality , Surveys and Questionnaires , Survival Analysis
9.
Clin Immunol ; 201: 30-34, 2019 04.
Article in English | MEDLINE | ID: mdl-30776520

ABSTRACT

Severe combined immunodeficiencies (SCID) comprise a group of genetic diseases characterized by abrogated development of T lymphocytes. In some case reports of atypical SCID patients elevated proportions of γδ T lymphocytes have been reported. However, it is unknown whether these γδ T cells modulate or reflect the patient's clinical phenotype. We investigated the frequency of elevated γδ T cell proportions and associations with clinical disease manifestations in a cohort of 76 atypical SCID patients. Increased proportions of γδ T lymphocytes were present in approximately 60% of these patients. Furthermore, we identified positive correlations between elevated proportions of γδ T cells and the occurrence of CMV infections and autoimmune cytopenias. We discuss that CMV infections might trigger an expansion of γδ T lymphocytes, which could drive the development of autoimmune cytopenias. We advocate that atypical SCID patients should be screened for elevated proportions of γδ T lymphocytes, CMV infection and autoimmune cytopenias.


Subject(s)
Cytomegalovirus Infections/immunology , Hematologic Diseases/immunology , Intraepithelial Lymphocytes/immunology , Severe Combined Immunodeficiency/immunology , Humans , Lymphocyte Count
10.
Allergy ; 74(9): 1691-1702, 2019 09.
Article in English | MEDLINE | ID: mdl-30793327

ABSTRACT

BACKGROUND: Pulmonary complications are responsible for high morbidity and mortality rates in patients with the rare immunodeficiency disorder STAT3 hyper-IgE syndrome (STAT3-HIES). The aim of this study was to expand knowledge about lung disease in STAT3-HIES. METHODS: The course of pulmonary disease, radiological and histopathological interrelations, therapeutic management, and the outcome of 14 STAT3-HIES patients were assessed. RESULTS: The patients' quality of life was compromised most by pulmonary disease. All 14 patients showed first signs of lung disease at a median onset of 1.5 years of age. Lung function revealed a mixed obstructive-restrictive impairment with reduced FEV1 and FVC in 75% of the patients. The severity of lung function impairment was associated with Aspergillus fumigatus infection and prior lung surgery. Severe lung tissue damage, with reduced numbers of ATP-binding cassette sub-family A member 3 (ABCA3) positive type II pneumocytes, was observed in the histological assessment of two deceased patients. Imaging studies of all patients above 6 years of age showed severe airway and parenchyma destruction. Lung surgeries frequently led to complications, including fistula formation. Long-term antifungal and antibacterial treatment proved to be beneficial, as were inhalation therapy, chest physiotherapy, and exercise. Regular immunoglobulin replacement therapy tended to stabilize lung function. CONCLUSIONS: Due to its severity, pulmonary disease in STAT3-HIES patients requires strict monitoring and intensive therapy.


Subject(s)
Disease Susceptibility , Job Syndrome/complications , Job Syndrome/metabolism , Lung Diseases/etiology , Lung Diseases/therapy , STAT3 Transcription Factor/metabolism , Adolescent , Adult , Anti-Infective Agents/therapeutic use , Biopsy , Child , Combined Modality Therapy , Disease Management , Female , Humans , Immunohistochemistry , Job Syndrome/genetics , Job Syndrome/mortality , Lung Diseases/diagnosis , Male , Middle Aged , Prognosis , Radiography, Thoracic , Respiratory Function Tests , STAT3 Transcription Factor/genetics , Symptom Assessment , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
11.
J Allergy Clin Immunol ; 139(4): 1302-1310.e4, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27658761

ABSTRACT

BACKGROUND: Absent T-cell immunity resulting in life-threatening infections provides a clear rationale for hematopoetic stem cell transplantation (HSCT) in patients with severe combined immunodeficiency (SCID). Combined immunodeficiencies (CIDs) and "atypical" SCID show reduced, not absent T-cell immunity. If associated with infections or autoimmunity, they represent profound combined immunodeficiency (P-CID), for which outcome data are insufficient for unambiguous early transplant decisions. OBJECTIVES: We sought to compare natural histories of severity-matched patients with/without subsequent transplantation and to determine whether immunologic and/or clinical parameters may be predictive for outcome. METHODS: In this prospective and retrospective observational study, we recruited nontransplanted patients with P-CID aged 1 to 16 years to compare natural histories of severity-matched patients with/without subsequent transplantation and to determine whether immunologic and/or clinical parameters may be predictive for outcome. RESULTS: A total of 51 patients were recruited (median age, 9.6 years). Thirteen of 51 had a genetic diagnosis of "atypical" SCID and 14 of 51 of CID. About half of the patients had less than 10% naive T cells, reduced/absent T-cell proliferation, and at least 1 significant clinical event/year, demonstrating their profound immunodeficiency. Nineteen patients (37%) underwent transplantation within 1 year of enrolment, and 5 of 51 patients died. Analysis of the HSCT decisions revealed the anticipated heterogeneity, favoring an ongoing prospective matched-pair analysis of patients with similar disease severity with or without transplantation. Importantly, so far neither the genetic diagnosis nor basic measurements of T-cell immunity were good predictors of disease evolution. CONCLUSIONS: The P-CID study for the first time characterizes a group of patients with nontypical SCID T-cell deficiencies from a therapeutic perspective. Because genetic and basic T-cell parameters provide limited guidance, prospective data from this study will be a helpful resource for guiding the difficult HSCT decisions in patients with P-CID.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , Severe Combined Immunodeficiency/therapy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Prospective Studies , Research Design
13.
Rheumatology (Oxford) ; 56(9): 1597-1606, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28859329

ABSTRACT

Objectives: The aim was to analyse factors influencing the individual colchicine dose in children with FMF, to evaluate the impact of dose adjustment on the clinical course and inflammation and to identify clinical parameters and biomarkers that predict dose increase in the near future. Methods: Data from 409 paediatric FMF patients (4566 visits) derived from the national auto-inflammatory diseases registry were analysed. Serum concentrations of S100 molecules were determined by ELISA. Results: The age-dependent colchicine dose is influenced by the present genotype. The body surface area is the anthropometric parameter that correlates best with the applied dosages. Colchicine introduction and dose increase lead to significant reduction of clinical symptoms and inflammation. During established colchicine therapy, an increase of one single biomarker increases the likelihood of a dose increment in the next 12 months with a factor of 1.62-1.94. A combination of biomarkers including S100 molecules increases this odds ratio up to 4.66 when analysing all patients and up to 7.27 when analysing patients with a high risk of severe disease. Conclusion: Colchicine therapy is currently guided mainly by the occurrence of clinical symptoms and serological inflammation. Other factors, such as the genotype, the body surface area and biomarkers, will help to manage colchicine therapy in a more individualized fashion. The additional analysis of S100 molecules as sensitive biomarkers will help to identify patients at risk for dose increases in the near future.


Subject(s)
Colchicine/administration & dosage , Familial Mediterranean Fever/drug therapy , Adolescent , Age Factors , Anthropometry/methods , Biomarkers/blood , Body Surface Area , Child , Child, Preschool , Colchicine/adverse effects , Colchicine/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/genetics , Female , Genotype , Humans , Male , Prognosis , Registries , S100 Proteins/blood , Severity of Illness Index
15.
J Clin Immunol ; 36(1): 73-84, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26604104

ABSTRACT

PURPOSE: Gain-of-function (GOF) mutations in the signal transducer and activator of transcription 1 (STAT1) result in unbalanced STAT signaling and cause immune dysregulation and immunodeficiency. The latter is often characterized by the susceptibility to recurrent Candida infections, resulting in the clinical picture of chronic mucocutaneous candidiasis (CMC). This study aims to assess the frequency of GOF STAT1 mutations in a large international cohort of CMC patients. METHODS: STAT1 was sequenced in genomic DNA from 57 CMC patients and 35 healthy family members. The functional relevance of nine different STAT1 variants was shown by flow cytometric analysis of STAT1 phosphorylation in patients' peripheral blood cells (PBMC) after stimulation with interferon (IFN)-α, IFN-γ or interleukin-27 respectively. Extended clinical data sets were collected and summarized for 26 patients. RESULTS: Heterozygous mutations within STAT1 were identified in 35 of 57 CMC patients (61%). Out of 39 familial cases from 11 families, 26 patients (67%) from 9 families and out of 18 sporadic cases, 9 patients (50%) were shown to have heterozygous mutations within STAT1. Thirteen distinct STAT1 mutations are reported in this paper. Eight of these mutations are known to cause CMC (p.M202V, p.A267V, p.R274W, p.R274Q, p.T385M, p.K388E, p.N397D, and p.F404Y). However, five STAT1 variants (p.F172L, p.Y287D, p.P293S, p.T385K and p.S466R) have not been reported before in CMC patients. CONCLUSION: STAT1 mutations are frequently observed in patients suffering from CMC. Thus, sequence analysis of STAT1 in CMC patients is advised. Measurement of IFN- or IL-induced STAT1 phosphorylation in PBMC provides a fast and reliable diagnostic tool and should be carried out in addition to genetic testing.


Subject(s)
Candidiasis, Chronic Mucocutaneous/diagnosis , Immunologic Deficiency Syndromes/diagnosis , Leukocytes, Mononuclear/immunology , Mutation/genetics , STAT1 Transcription Factor/metabolism , Adult , Candidiasis, Chronic Mucocutaneous/genetics , Cells, Cultured , Cytokines/metabolism , DNA Mutational Analysis , Female , Humans , Immunologic Deficiency Syndromes/genetics , Male , Pedigree , Phenotype , Protein Structure, Tertiary/genetics , STAT1 Transcription Factor/genetics
16.
Pediatr Allergy Immunol ; 27(2): 177-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26592211

ABSTRACT

BACKGROUND: Hyper-IgE syndromes (HIES) are primary immunodeficiency disorders characterized by elevated serum IgE, eczema, and recurrent infections. Despite the availability of confirmatory molecular diagnosis of several distinct HIES entities, the differentiation of HIES particularly from severe forms of atopic dermatitis remains a challenge. The two most common forms of HIES are caused by mutations in the genes STAT3 and DOCK8. METHODS: Here, we assess the clinical and immunologic phenotype of DOCK8- and STAT3-HIES patients including the cell activation, proliferation, and cytokine release after stimulation. RESULTS: Existing HIES scoring systems are helpful to identify HIES patients. However, those scores may fail in infants and young children due to the age-related lack of clinical symptoms. Furthermore, our long-term observations showed a striking variation of laboratory results over time in the individual patient. Reduced memory B-cell counts in concert with low specific antibody production are the most consistent findings likely contributing to the high susceptibility to bacterial and fungal infection. In DOCK8-HIES, T-cell lymphopenia and low IFN-gamma secretion after stimulation were common, likely promoting viral infections. In contrast to STAT3-HIES, DOCK8-HIES patients showed more severe inflammation with regard to allergic manifestations, elevated activation markers (HLA-DR, CD69, CD86, and CD154), and significantly increased inflammatory cytokines (IL1-beta, IL4, IL6, and IFN-gamma). CONCLUSION: Differentiating HIES from other diseases such as atopic dermatitis early in life is essential for patients because treatment modalities differ. To expedite the diagnosis process, we propose here a diagnostic workflow.


Subject(s)
Dermatitis, Atopic/diagnosis , Guanine Nucleotide Exchange Factors/genetics , Job Syndrome/diagnosis , Mutation/genetics , STAT3 Transcription Factor/genetics , B-Lymphocytes/immunology , Cells, Cultured , Child, Preschool , Cytokines/metabolism , Diagnosis, Differential , Female , Humans , Immunoglobulin E/blood , Immunologic Memory , Infant , Job Syndrome/genetics , Lymphocyte Activation/genetics , Male , T-Lymphocytes/immunology
17.
J Allergy Clin Immunol ; 136(3): 703-712.e10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25843314

ABSTRACT

BACKGROUND: The clinical and immunologic features of CD27 deficiency remain obscure because only a few patients have been identified to date. OBJECTIVE: We sought to identify novel mutations in TNFRSF7/CD27 and to provide an overview of clinical, immunologic, and laboratory phenotypes in patients with CD27 deficiency. METHODS: Review of the medical records and molecular, genetic, and flow cytometric analyses of the patients and family members were performed. Treatment outcomes of previously described patients were followed up. RESULTS: In addition to the previously reported homozygous mutations c.G24A/p.W8X (n = 2) and c.G158A/p.C53Y (n = 8), 4 novel mutations were identified: homozygous missense c.G287A/p.C96Y (n = 4), homozygous missense c.C232T/p.R78W (n = 1), heterozygous nonsense c.C30A/p.C10X (n = 1), and compound heterozygous c.C319T/p.R107C-c.G24A/p.W8X (n = 1). EBV-associated lymphoproliferative disease/hemophagocytic lymphohistiocytosis, Hodgkin lymphoma, uveitis, and recurrent infections were the predominant clinical features. Expression of cell-surface and soluble CD27 was significantly reduced in patients and heterozygous family members. Immunoglobulin substitution therapy was administered in 5 of the newly diagnosed cases. CONCLUSION: CD27 deficiency is potentially fatal and should be excluded in all cases of severe EBV infections to minimize diagnostic delay. Flow cytometric immunophenotyping offers a reliable initial test for CD27 deficiency. Determining the precise role of CD27 in immunity against EBV might provide a framework for new therapeutic concepts.


Subject(s)
Epstein-Barr Virus Infections/genetics , Hodgkin Disease/genetics , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphoproliferative Disorders/genetics , Mutation , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Uveitis/genetics , Adolescent , Child, Preschool , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Exome , Female , Flow Cytometry , Heterozygote , Hodgkin Disease/diagnosis , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Homozygote , Humans , Immunophenotyping , Infant , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/pathology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Uveitis/diagnosis , Uveitis/immunology , Uveitis/pathology , Young Adult
18.
J Allergy Clin Immunol ; 136(2): 402-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25724123

ABSTRACT

BACKGROUND: Mutations in dedicator of cytokinesis 8 (DOCK8) cause a combined immunodeficiency (CID) also classified as autosomal recessive (AR) hyper-IgE syndrome (HIES). Recognizing patients with CID/HIES is of clinical importance because of the difference in prognosis and management. OBJECTIVES: We sought to define the clinical features that distinguish DOCK8 deficiency from other forms of HIES and CIDs, study the mutational spectrum of DOCK8 deficiency, and report on the frequency of specific clinical findings. METHODS: Eighty-two patients from 60 families with CID and the phenotype of AR-HIES with (64 patients) and without (18 patients) DOCK8 mutations were studied. Support vector machines were used to compare clinical data from 35 patients with DOCK8 deficiency with those from 10 patients with AR-HIES without a DOCK8 mutation and 64 patients with signal transducer and activator of transcription 3 (STAT3) mutations. RESULTS: DOCK8-deficient patients had median IgE levels of 5201 IU, high eosinophil levels of usually at least 800/µL (92% of patients), and low IgM levels (62%). About 20% of patients were lymphopenic, mainly because of low CD4(+) and CD8(+) T-cell counts. Fewer than half of the patients tested produced normal specific antibody responses to recall antigens. Bacterial (84%), viral (78%), and fungal (70%) infections were frequently observed. Skin abscesses (60%) and allergies (73%) were common clinical problems. In contrast to STAT3 deficiency, there were few pneumatoceles, bone fractures, and teething problems. Mortality was high (34%). A combination of 5 clinical features was helpful in distinguishing patients with DOCK8 mutations from those with STAT3 mutations. CONCLUSIONS: DOCK8 deficiency is likely in patients with severe viral infections, allergies, and/or low IgM levels who have a diagnosis of HIES plus hypereosinophilia and upper respiratory tract infections in the absence of parenchymal lung abnormalities, retained primary teeth, and minimal trauma fractures.


Subject(s)
Bacterial Infections/complications , Guanine Nucleotide Exchange Factors/deficiency , Job Syndrome/complications , Phenotype , Skin Diseases/complications , Virus Diseases/complications , Adolescent , Adult , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Antigens, Viral/blood , Antigens, Viral/immunology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/mortality , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Eosinophils/immunology , Eosinophils/pathology , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin E/genetics , Immunoglobulin M/blood , Immunoglobulin M/genetics , Infant , Job Syndrome/genetics , Job Syndrome/immunology , Job Syndrome/mortality , Lymphocyte Count , Male , Middle Aged , Mutation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/mortality , Support Vector Machine , Survival Analysis , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/mortality
19.
Pediatr Blood Cancer ; 62(6): 1091-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25504888

ABSTRACT

Mutations in SLC46A1 result in a defect of the proton coupled folate transporter (PCFT) and are the basis of hereditary folate malabsorption (HFM). Patients with HFM frequently present with neurodevelopmental delay and megaloblastic anemia. Some cases may be complicated by additional lymphopenia and immunodeficiency. We report a patient with a new homozygous mutation in the SLC46A1 gene. The boy presented with early-onset pancytopenia and secondary immunodeficiency. We provide clinical and molecular observations that extend the phenotypic description of HFM and highlight diagnostic as well as therapeutic pitfalls in this rare condition.


Subject(s)
Folic Acid Deficiency/complications , Immunologic Deficiency Syndromes/etiology , Malabsorption Syndromes/complications , Pancytopenia/etiology , Adolescent , Adult , Child , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL