Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
3.
Blood ; 143(10): 872-881, 2024 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-37992218

RÉSUMÉ

ABSTRACT: Primary hemophagocytic lymphohistiocytosis (pHLH) is a life-threatening hyperinflammatory syndrome that develops mainly in patients with genetic disorders of lymphocyte cytotoxicity and X-linked lymphoproliferative syndromes. Previous studies with etoposide-based treatment followed by hematopoetic stem cell transplantation (HSCT) resulted in 5-year survival of 50% to 59%. Contemporary data are lacking. We evaluated 88 patients with pHLH documented in the international HLH registry from 2016-2021. In 12 of 88 patients, diagnosis was made without HLH activity, based on siblings or albinism. Major HLH-directed drugs (etoposide, antithymocyte globulin, alemtuzumab, emapalumab, ruxolitinib) were administered to 66 of 76 patients who were symptomatic (86% first-line etoposide); 16 of 57 patients treated with etoposide and 3 of 9 with other first-line treatment received salvage therapy. HSCT was performed in 75 patients; 7 patients died before HSCT. Three-year probability of survival (pSU) was 82% (confidence interval [CI], 72%-88%) for the entire cohort and 77% (CI, 64%-86%) for patients receiving first-line etoposide. Compared with the HLH-2004 study, both pre-HSCT and post-HSCT survival of patients receiving first-line etoposide improved, 83% to 91% and 70% to 88%. Differences to HLH-2004 included preferential use of reduced-toxicity conditioning and reduced time from diagnosis to HSCT (from 148 to 88 days). Three-year pSU was lower with haploidentical (4 of 9 patients [44%]) than with other donors (62 of 66 [94%]; P < .001). Importantly, early HSCT for patients who were asymptomatic resulted in 100% survival, emphasizing the potential benefit of newborn screening. This contemporary standard-of-care study of patients with pHLH reveals that first-line etoposide-based therapy is better than previously reported, providing a benchmark for novel treatment regimes.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Lymphohistiocytose hémophagocytaire , Syndromes lymphoprolifératifs , Nouveau-né , Humains , Étoposide/usage thérapeutique , Lymphohistiocytose hémophagocytaire/traitement médicamenteux , Lymphohistiocytose hémophagocytaire/diagnostic , Résultat thérapeutique , Transplantation de cellules souches hématopoïétiques/méthodes , Syndromes lymphoprolifératifs/étiologie
5.
J Exp Med ; 220(5)2023 05 01.
Article de Anglais | MEDLINE | ID: mdl-36880831

RÉSUMÉ

X-linked recessive deficiency of TLR7, a MyD88- and IRAK-4-dependent endosomal ssRNA sensor, impairs SARS-CoV-2 recognition and type I IFN production in plasmacytoid dendritic cells (pDCs), thereby underlying hypoxemic COVID-19 pneumonia with high penetrance. We report 22 unvaccinated patients with autosomal recessive MyD88 or IRAK-4 deficiency infected with SARS-CoV-2 (mean age: 10.9 yr; 2 mo to 24 yr), originating from 17 kindreds from eight countries on three continents. 16 patients were hospitalized: six with moderate, four with severe, and six with critical pneumonia, one of whom died. The risk of hypoxemic pneumonia increased with age. The risk of invasive mechanical ventilation was also much greater than in age-matched controls from the general population (OR: 74.7, 95% CI: 26.8-207.8, P < 0.001). The patients' susceptibility to SARS-CoV-2 can be attributed to impaired TLR7-dependent type I IFN production by pDCs, which do not sense SARS-CoV-2 correctly. Patients with inherited MyD88 or IRAK-4 deficiency were long thought to be selectively vulnerable to pyogenic bacteria, but also have a high risk of hypoxemic COVID-19 pneumonia.


Sujet(s)
COVID-19 , Facteur de différenciation myéloïde-88 , Enfant , Humains , Protéines adaptatrices de la transduction du signal , COVID-19/complications , Facteur de différenciation myéloïde-88/génétique , SARS-CoV-2 , Récepteur de type Toll-7
6.
J Clin Immunol ; 43(5): 965-978, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36843153

RÉSUMÉ

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.


Sujet(s)
Lymphopénie , Immunodéficience combinée grave , Enfant , Nouveau-né , Humains , Dépistage néonatal/méthodes , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/épidémiologie , Immunodéficience combinée grave/thérapie , Études prospectives , Lymphopénie/diagnostic , ADN , Allemagne/épidémiologie , Récepteurs aux antigènes des cellules T/génétique
7.
Leukemia ; 37(3): 505-517, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36707619

RÉSUMÉ

Treatment of chronic myeloid leukemia has improved significantly with the introduction of tyrosine kinase inhibitors (TKIs), and treatment guidelines based on numerous clinical trials are available for chronic phase disease. However for CML in the blast phase (CML-BP), prognosis remains poor and treatment options are much more limited. The spectrum of treatment strategies for children and adolescents with CML-BP has largely evolved empirically and includes treatment principles derived from adult CML-BP and pediatric acute leukemia. Given this heterogeneity of treatment approaches, we formed an international panel of pediatric CML experts to develop recommendations for consistent therapy in children and adolescents with this high-risk disease based on the current literature and national standards. Recommendations include detailed information on initial diagnosis and treatment monitoring, differentiation from Philadelphia-positive acute leukemia, subtype-specific selection of induction therapy, and combination with tyrosine kinase inhibitors. Given that allogeneic hematopoietic stem cell transplantation currently remains the primary curative intervention for CML-BP, we also provide recommendations for the timing of transplantation, donor and graft selection, selection of a conditioning regimen and prophylaxis for graft-versus-host disease, post-transplant TKI therapy, and management of molecular relapse. Management according to the treatment recommendations presented here is intended to provide the basis for the design of future prospective clinical trials to improve outcomes for this challenging disease.


Sujet(s)
Maladie du greffon contre l'hôte , Transplantation de cellules souches hématopoïétiques , Leucémie myéloïde chronique BCR-ABL positive , Leucémie aigüe myéloïde , Adulte , Humains , Enfant , Adolescent , Crise blastique/thérapie , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/diagnostic , Pronostic
8.
J Allergy Clin Immunol ; 149(1): 388-399.e4, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34033843

RÉSUMÉ

BACKGROUND: Rubella virus-induced granulomas have been described in patients with various inborn errors of immunity. Most defects impair T-cell immunity, suggesting a critical role of T cells in rubella elimination. However, the molecular mechanism of virus control remains elusive. OBJECTIVE: This study sought to understand the defective effector mechanism allowing rubella vaccine virus persistence in granulomas. METHODS: Starting from an index case with Griscelli syndrome type 2 and rubella skin granulomas, this study combined an international survey with a literature search to identify patients with cytotoxicity defects and granuloma. The investigators performed rubella virus immunohistochemistry and PCR and T-cell migration assays. RESULTS: This study identified 21 patients with various genetically confirmed cytotoxicity defects, who presented with skin and visceral granulomas. Rubella virus was demonstrated in all 12 accessible biopsies. Granuloma onset was typically before 2 years of age and lesions persisted from months to years. Granulomas were particularly frequent in MUNC13-4 and RAB27A deficiency, where 50% of patients at risk were affected. Although these proteins have also been implicated in lymphocyte migration, 3-dimensional migration assays revealed no evidence of impaired migration of patient T cells. Notably, patients showed no evidence of reduced control of concomitantly given measles, mumps, or varicella live-attenuated vaccine or severe infections with other viruses. CONCLUSIONS: This study identified lymphocyte cytotoxicity as a key effector mechanism for control of rubella vaccine virus, without evidence for its need in control of live measles, mumps, or varicella vaccines. Rubella vaccine-induced granulomas are a novel phenotype with incomplete penetrance of genetic disorders of cytotoxicity.


Sujet(s)
Granulome/étiologie , Vaccin antirubéoleux/effets indésirables , Lymphocytes T/immunologie , Enfant , Enfant d'âge préscolaire , Femelle , Granulome/génétique , Granulome/immunologie , Granulome/virologie , Humains , Nourrisson , Phénotype , Rubéole/génétique , Rubéole/immunologie , Rubéole/virologie , Peau/immunologie , Peau/virologie
9.
J Inherit Metab Dis ; 44(6): 1441-1452, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34389986

RÉSUMÉ

Fucosylation is essential for intercellular and intracellular recognition, cell-cell interaction, fertilization, and inflammatory processes. Only five types of congenital disorders of glycosylation (CDG) related to an impaired fucosylation have been described to date: FUT8-CDG, FCSK-CDG, POFUT1-CDG SLC35C1-CDG, and the only recently described GFUS-CDG. This review summarizes the clinical findings of all hitherto known 25 patients affected with those defects with regard to their pathophysiology and genotype. In addition, we describe five new patients with novel variants in the SLC35C1 gene. Furthermore, we discuss the efficacy of fucose therapy approaches within the different defects.


Sujet(s)
Troubles congénitaux de la glycosylation/traitement médicamenteux , Troubles congénitaux de la glycosylation/génétique , Fucose/usage thérapeutique , Transporteurs de monosaccharides/génétique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Femelle , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Glycoprotéines , Glycosylation , Humains , Nourrisson , Mâle , Résultat thérapeutique , Jeune adulte
10.
J Exp Med ; 218(2)2021 02 01.
Article de Anglais | MEDLINE | ID: mdl-33170215

RÉSUMÉ

The identification and characterization of rare immune cell populations in humans can be facilitated by their growth advantage in the context of specific genetic diseases. Here, we use autoimmune lymphoproliferative syndrome to identify a population of FAS-controlled TCRαß+ T cells. They include CD4+, CD8+, and double-negative T cells and can be defined by a CD38+CD45RA+T-BET- expression pattern. These unconventional T cells are present in healthy individuals, are generated before birth, are enriched in lymphoid tissue, and do not expand during acute viral infection. They are characterized by a unique molecular signature that is unambiguously different from other known T cell differentiation subsets and independent of CD4 or CD8 expression. Functionally, FAS-controlled T cells represent highly proliferative, noncytotoxic T cells with an IL-10 cytokine bias. Mechanistically, regulation of this physiological population is mediated by FAS and CTLA4 signaling, and its survival is enhanced by mTOR and STAT3 signals. Genetic alterations in these pathways result in expansion of FAS-controlled T cells, which can cause significant lymphoproliferative disease.


Sujet(s)
Antigènes CD38/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Antigènes CD45/métabolisme , Antigènes CD95/immunologie , Adolescent , Adulte , Sujet âgé , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Nouveau-né , Activation des lymphocytes/immunologie , Syndromes lymphoprolifératifs/immunologie , Mâle , Adulte d'âge moyen , Transduction du signal/immunologie , Jeune adulte
11.
J Clin Immunol ; 40(5): 708-717, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32458183

RÉSUMÉ

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.


Sujet(s)
Immunodéficience combinée grave/épidémiologie , Femelle , Allemagne/épidémiologie , Humains , Incidence , Nourrisson , Nouveau-né , Mâle , Dépistage néonatal , Phénotype , Immunodéficience combinée grave/mortalité , Enquêtes et questionnaires , Analyse de survie
12.
Front Immunol ; 10: 2481, 2019.
Article de Anglais | MEDLINE | ID: mdl-31695698

RÉSUMÉ

Neutrophils respond to various stimuli by decondensing and releasing nuclear chromatin characterized by citrullinated histones as neutrophil extracellular traps (NETs). This achieves pathogen immobilization or initiation of thrombosis, yet the molecular mechanisms of NET formation remain elusive. Peptidyl arginine deiminase-4 (PAD4) achieves protein citrullination and has been intricately linked to NET formation. Here we show that citrullination represents a major regulator of proteolysis in the course of NET formation. Elevated cytosolic calcium levels trigger both peptidylarginine deiminase-4 (PAD4) and calpain activity in neutrophils resulting in nuclear decondensation typical of NETs. Interestingly, PAD4 relies on proteolysis by calpain to achieve efficient nuclear lamina breakdown and chromatin decondensation. Pharmacological or genetic inhibition of PAD4 and calpain strongly inhibit chromatin decondensation of human and murine neutrophils in response to calcium ionophores as well as the proteolysis of nuclear proteins like lamin B1 and high mobility group box protein 1 (HMGB1). Taken together, the concerted action of PAD4 and calpain induces nuclear decondensation in the course of calcium-mediated NET formation.


Sujet(s)
Calpain/immunologie , Citrullination/immunologie , Pièges extracellulaires/immunologie , Granulocytes neutrophiles/immunologie , Lamina nucléaire/immunologie , Animaux , Calpain/génétique , Citrullination/génétique , Pièges extracellulaires/génétique , Humains , Souris , Souris knockout , Granulocytes neutrophiles/cytologie , Lamina nucléaire/génétique , Protein-arginine deiminase Type 4/génétique , Protein-arginine deiminase Type 4/immunologie
13.
J Invest Dermatol ; 136(11): 2150-2157, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27375110

RÉSUMÉ

Severe generalized junctional epidermolysis bullosa, a lethal hereditary blistering disorder, is usually treated by palliative care. Allogeneic stem cell transplantation (SCT) has been proposed as a therapeutic approach, yet without clinical evidence. Decision making was evaluated retrospectively in 76 patients with severe generalized junctional epidermolysis bullosa born in the years 2000-2015. The diagnosis was based on the absence of laminin-332 in skin biopsies. With an incidence of 1 of 150,000, severe generalized junctional epidermolysis bullosa occurred more often than published previously. Eleven as yet unreported mutations in the laminin-332 genes were detected. Although patients homozygous for the LAMB3 mutation c.1903C>T lived longer than the others, life expectancy was greatly diminished (10.8 vs. 4.6 months). Most patients failed to thrive. In two patients with initially normal weight gain, the decision for SCT from haploidentical bone marrow or peripheral blood was made. Despite transiently increasing skin erosions, the clinical status of both subjects stabilized for several weeks after SCT, but finally deteriorated. Graft cells, but no laminin-332, were detected in skin biopsies. The patients died 96 and 129 days after SCT, respectively, one of them after receiving additional skin grafts. Treatment of severe generalized junctional epidermolysis bullosa by SCT is a last-ditch attempt still lacking proof of efficacy.


Sujet(s)
Molécules d'adhérence cellulaire/génétique , ADN/génétique , Épidermolyse bulleuse jonctionnelle/génétique , Laminine/génétique , Mutation , Transplantation de cellules souches/méthodes , Biopsie , Molécules d'adhérence cellulaire/métabolisme , Analyse de mutations d'ADN , Prise de décision , Épidermolyse bulleuse jonctionnelle/diagnostic , Épidermolyse bulleuse jonctionnelle/thérapie , Femelle , Études de suivi , Génotype , Homozygote , Humains , Nourrisson , Laminine/métabolisme , Mâle , Études rétrospectives , Peau/métabolisme , Peau/anatomopathologie ,
14.
Blood ; 128(2): 227-38, 2016 07 14.
Article de Anglais | MEDLINE | ID: mdl-27099149

RÉSUMÉ

Autoimmune lymphoproliferative syndrome (ALPS) is a human disorder characterized by defective Fas signaling, resulting in chronic benign lymphoproliferation and accumulation of TCRαß(+) CD4(-) CD8(-) double-negative T (DNT) cells. Although their phenotype resembles that of terminally differentiated or exhausted T cells, lack of KLRG1, high eomesodermin, and marginal T-bet expression point instead to a long-lived memory state with potent proliferative capacity. Here we show that despite their terminally differentiated phenotype, human ALPS DNT cells exhibit substantial mitotic activity in vivo. Notably, hyperproliferation of ALPS DNT cells is associated with increased basal and activation-induced phosphorylation of serine-threonine kinases Akt and mechanistic target of rapamycin (mTOR). The mTOR inhibitor rapamycin abrogated survival and proliferation of ALPS DNT cells, but not of CD4(+) or CD8(+) T cells in vitro. In vivo, mTOR inhibition reduced proliferation and abnormal differentiation by DNT cells. Importantly, increased mitotic activity and hyperactive mTOR signaling was also observed in recently defined CD4(+) or CD8(+) precursor DNT cells, and mTOR inhibition specifically reduced these cells in vivo, indicating abnormal programming of Fas-deficient T cells before the DNT stage. Thus, our results identify the mTOR pathway as a major regulator of lymphoproliferation and aberrant differentiation in ALPS.


Sujet(s)
Syndrome lymphoprolifératif avec auto-immunité/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Différenciation cellulaire/immunologie , Transduction du signal/immunologie , Sérine-thréonine kinases TOR/immunologie , Adolescent , Adulte , Syndrome lymphoprolifératif avec auto-immunité/anatomopathologie , Lymphocytes T CD4+/anatomopathologie , Lymphocytes T CD8+/anatomopathologie , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Lectines de type C/immunologie , Antigènes CD45/immunologie , Mâle , Protéines proto-oncogènes c-akt/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs immunologiques , Transactivateurs/immunologie
16.
Pediatrics ; 125(1): e169-73, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-20026502

RÉSUMÉ

Autoimmune lymphoproliferative syndrome (ALPS) is an uncommon disorder of Fas-mediated apoptosis that results in impaired lymphocyte death and, therefore, disturbed immune homeostasis. Besides presentation with lymphadenopathy and splenomegaly, patients with ALPS have a high incidence of autoimmune phenomena. To our knowledge, this is the first description of polyarteritis nodosa that includes numerous arterial aneurysms in a child with ALPS. Active vasculitis resolved after allogeneic hematopoietic stem cell transplantation. This report of polyarteritis nodosa associated with human ALPS supports previous findings in Fas-deficient mouse models that frequently develop vasculitic manifestations and suggests that apoptotic defects of lymphocytes may play a role in the pathophysiology of systemic vasculitis. Thus, patients with ALPS might be more susceptible to autoimmune vessel inflammation. This case furthermore emphasizes that even rare autoimmune manifestations should be considered and investigated in patients with immunodeficiencies, because that might help in planning treatment strategies for these patients.


Sujet(s)
Syndrome lymphoprolifératif avec auto-immunité/complications , Syndrome lymphoprolifératif avec auto-immunité/diagnostic , Transplantation de cellules souches hématopoïétiques/méthodes , Polyartérite noueuse/complications , Polyartérite noueuse/diagnostic , Syndrome lymphoprolifératif avec auto-immunité/chirurgie , Études de suivi , Humains , Nourrisson , Angiographie par résonance magnétique , Mâle , Monitorage physiologique/méthodes , Polyartérite noueuse/chirurgie , Maladies rares , Appréciation des risques , Tomodensitométrie , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE