Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 708
Filter
1.
J Clin Immunol ; 44(5): 116, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38676861

ABSTRACT

PURPOSE: A patient with X-linked agammaglobulinemia (XLA) and severe tick-borne encephalitis (TBE) was treated with TBE virus (TBEV) IgG positive plasma. The patient's clinical response, humoral and cellular immune responses were characterized pre- and post-infection. METHODS: ELISA and neutralisation assays were performed on sera and TBEV PCR assay on sera and cerebrospinal fluid. T cell assays were conducted on peripheral blood the patient and five healthy vaccinated controls. RESULTS: The patient was admitted to the hospital with headache and fever. He was not vaccinated against TBE but receiving subcutaneous IgG-replacement therapy (IGRT). TBEV IgG antibodies were low-level positive (due to scIGRT), but the TBEV IgM and TBEV neutralisation tests were negative. During hospitalisation his clinical condition deteriorated (Glasgow coma scale 3/15) and he was treated in the ICU with corticosteroids and external ventricular drainage. He was then treated with plasma containing TBEV IgG without apparent side effects. His symptoms improved within a few days and the TBEV neutralisation test converted to positive. Robust CD8+ T cell responses were observed at three and 18-months post-infection, in the absence of B cells. This was confirmed by tetramers specific for TBEV. CONCLUSION: TBEV IgG-positive plasma given to an XLA patient with TBE without evident adverse reactions may have contributed to a positive clinical outcome. Similar approaches could offer a promising foundation for researching therapeutic options for patients with humoral immunodeficiencies. Importantly, a robust CD8+ T cell response was observed after infection despite the lack of B cells and indicates that these patients can clear acute viral infections and could benefit from future vaccination programs.


Subject(s)
Agammaglobulinemia , Antibodies, Viral , Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Genetic Diseases, X-Linked , Immunoglobulin G , T-Lymphocytes , Humans , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/diagnosis , Encephalitis, Tick-Borne/therapy , Male , Agammaglobulinemia/immunology , Agammaglobulinemia/therapy , Encephalitis Viruses, Tick-Borne/immunology , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/therapy , Immunoglobulin G/blood , Immunoglobulin G/immunology , Antibodies, Viral/blood , T-Lymphocytes/immunology , Treatment Outcome , Adult , Immunization, Passive/methods
2.
Nat Commun ; 15(1): 3662, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688902

ABSTRACT

Hematopoietic stem cell gene therapy (GT) using a γ-retroviral vector (γ-RV) is an effective treatment for Severe Combined Immunodeficiency due to Adenosine Deaminase deficiency. Here, we describe a case of GT-related T-cell acute lymphoblastic leukemia (T-ALL) that developed 4.7 years after treatment. The patient underwent chemotherapy and haploidentical transplantation and is currently in remission. Blast cells contain a single vector insertion activating the LIM-only protein 2 (LMO2) proto-oncogene, confirmed by physical interaction, and low Adenosine Deaminase (ADA) activity resulting from methylation of viral promoter. The insertion is detected years before T-ALL in multiple lineages, suggesting that further hits occurred in a thymic progenitor. Blast cells contain known and novel somatic mutations as well as germline mutations which may have contributed to transformation. Before T-ALL onset, the insertion profile is similar to those of other ADA-deficient patients. The limited incidence of vector-related adverse events in ADA-deficiency compared to other γ-RV GT trials could be explained by differences in transgenes, background disease and patient's specific factors.


Subject(s)
Adenosine Deaminase , Agammaglobulinemia , Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Mas , Severe Combined Immunodeficiency , Humans , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Genetic Therapy/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Severe Combined Immunodeficiency/therapy , Severe Combined Immunodeficiency/genetics , Genetic Vectors/genetics , Agammaglobulinemia/therapy , Agammaglobulinemia/genetics , Male , Retroviridae/genetics
3.
J Clin Immunol ; 44(5): 107, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38676811

ABSTRACT

PURPOSE: Patients with adenosine deaminase 1 deficient severe combined immunodeficiency (ADA-SCID) are initially treated with enzyme replacement therapy (ERT) with polyethylene glycol-modified (PEGylated) ADA while awaiting definitive treatment with hematopoietic stem cell transplant (HSCT) or gene therapy. Beginning in 1990, ERT was performed with PEGylated bovine intestinal ADA (ADAGEN®). In 2019, a PEGylated recombinant bovine ADA (Revcovi®) replaced ADAGEN following studies in older patients previously treated with ADAGEN for many years. There are limited longitudinal data on ERT-naïve newborns treated with Revcovi. METHODS: We report our clinical experience with Revcovi as initial bridge therapy in three newly diagnosed infants with ADA-SCID, along with comprehensive biochemical and immunologic data. RESULTS: Revcovi was initiated at twice weekly dosing (0.2 mg/kg intramuscularly), and monitored by following plasma ADA activity and the concentration of total deoxyadenosine nucleotides (dAXP) in erythrocytes. All patients rapidly achieved a biochemically effective level of plasma ADA activity, and red cell dAXP were eliminated within 2-3 months. Two patients reconstituted B-cells and NK-cells within the first month of ERT, followed by naive T-cells one month later. The third patient reconstituted all lymphocyte subsets within the first month of ERT. One patient experienced declining lymphocyte counts with improvement following Revcovi dose escalation. Two patients developed early, self-resolving thrombocytosis, but no thromboembolic events occurred. CONCLUSION: Revcovi was safe and effective as initial therapy to restore immune function in these newly diagnosed infants with ADA-SCID, however, time course and degree of reconstitution varied. Revcovi dose may need to be optimized based on immune reconstitution, clinical status, and biochemical data.


Subject(s)
Adenosine Deaminase , Agammaglobulinemia , Enzyme Replacement Therapy , Severe Combined Immunodeficiency , Humans , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Severe Combined Immunodeficiency/therapy , Infant , Agammaglobulinemia/therapy , Male , Female , Infant, Newborn , Animals , Treatment Outcome , Immune Reconstitution , Recombinant Proteins/therapeutic use
5.
Int Arch Allergy Immunol ; 185(4): 370-381, 2024.
Article in English | MEDLINE | ID: mdl-38194943

ABSTRACT

INTRODUCTION: X-linked lymphoproliferative syndrome (XLP) is a rare primary immune deficiency. Two types of XLP have been described: XLP-1 and XLP-2. METHODS: We found 7 patients with XLP (3 had XLP-1 and 4 had XLP-2) after reviewing the data from Pediatric Immunodeficiency Clinic from 1997 to 2021. RESULTS: Mean age at diagnosis was 3.8 years, and mean delay in diagnosis was 2.6 years. Five patients had recurrent episodes of infections. Four patients developed at least one episode of hemophagocytic lymphohistiocytosis (HLH) (2 with XLP-1 and 2 with XLP-2). Of these, 2 had recurrent HLH (both with XLP-2). Epstein-Barr virus (EBV) infection was detected in 2 (1 with XLP-1 and 1 with XLP-2). Both these patients had HLH. One child with XLP-2 had inflammatory bowel disease. Hypogammaglobulinemia was seen in 3 (2 with XLP-1 and 1 with XLP-2). Genetic analysis showed previously reported variants in 5, while 2 had novel variants (one in exon 7 of XIAP gene [c.1370dup p.Asn457Lysfs Ter16] and other had splice site variant in intron 1 of SH2D1A gene [c.138-2_138-1insG]). Episodes of HLH were managed with intravenous immunoglobulin (IVIg), methylprednisolone, oral prednisolone, cyclosporine, and rituximab. Inflammatory bowel disease was managed using oral prednisolone and azathioprine. One patient underwent haploidentical hematopoietic stem cell transplantation. One child with XLP-2 and WAS died because of fulminant pneumonia. DISCUSSION/CONCLUSIONS: XLP should be considered as a strong possibility in any patient with features of HLH, repeated infections with hypogammaglobulinemia, persistent EBV infection, and early-onset IBD.


Subject(s)
Agammaglobulinemia , Epstein-Barr Virus Infections , Inflammatory Bowel Diseases , Lymphohistiocytosis, Hemophagocytic , Lymphoproliferative Disorders , Child , Humans , Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/therapy , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/therapy , Prednisolone
6.
Immunol Rev ; 322(1): 157-177, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38233996

ABSTRACT

Inborn errors of immunity (IEI) present a unique paradigm in the realm of gene therapy, emphasizing the need for precision in therapeutic design. As gene therapy transitions from broad-spectrum gene addition to careful modification of specific genes, the enduring safety and effectiveness of these therapies in clinical settings have become crucial. This review discusses the significance of IEIs as foundational models for pioneering and refining precision medicine. We explore the capabilities of gene addition and gene correction platforms in modifying the DNA sequence of primary cells tailored for IEIs. The review uses four specific IEIs to highlight key issues in gene therapy strategies: X-linked agammaglobulinemia (XLA), X-linked chronic granulomatous disease (X-CGD), X-linked hyper IgM syndrome (XHIGM), and immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX). We detail the regulatory intricacies and therapeutic innovations for each disorder, incorporating insights from relevant clinical trials. For most IEIs, regulated expression is a vital aspect of the underlying biology, and we discuss the importance of endogenous regulation in developing gene therapy strategies.


Subject(s)
Agammaglobulinemia , Genetic Diseases, X-Linked , Intestinal Diseases , Humans , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Intestinal Diseases/genetics , Intestinal Diseases/therapy , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Genetic Therapy
7.
J Allergy Clin Immunol Pract ; 12(5): 1139-1149, 2024 May.
Article in English | MEDLINE | ID: mdl-38246560

ABSTRACT

During the past 20 years, gene editing has emerged as a novel form of gene therapy. Since the publication of the first potentially therapeutic gene editing platform for genetic disorders, increasingly sophisticated editing technologies have been developed. As with viral vector-mediated gene addition, inborn errors of immunity are excellent candidate diseases for a corrective autologous hematopoietic stem cell gene editing strategy. Research on gene editing for inborn errors of immunity is still entirely preclinical, with no trials yet underway. However, with editing techniques maturing, scientists are investigating this novel form of gene therapy in context of an increasing number of inborn errors of immunity. Here, we present an overview of these studies and the recent progress moving these technologies closer to clinical benefit.


Subject(s)
Gene Editing , Genetic Therapy , Humans , Gene Editing/methods , Genetic Therapy/methods , Animals , CRISPR-Cas Systems , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Severe Combined Immunodeficiency/immunology , Hematopoietic Stem Cell Transplantation
8.
J Allergy Clin Immunol Pract ; 12(3): 744-752.e3, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38122866

ABSTRACT

BACKGROUND: Good's syndrome (GS) is a rare immune deficiency described almost 6 decades ago. Despite numerous published individual case reports and data collected in cross-sectional studies of small cohorts, the natural history and long-term outcomes of this disease remain unknown. OBJECTIVE: We aimed to determine the clinical and laboratory evolution of 8 adults diagnosed with GS and consecutively evaluated between 1983 and 2023. METHODS: In this prospective, longitudinal cohort study, newly diagnosed patients with GS were followed through repeated measures of clinical, immune, and hematologic changes, as well as targeted genetic screening. RESULTS: All patients reported a healthy childhood and adolescence with symptom onset during the third or fourth decade of life. All presented to our center with recurrent bacterial sinopulmonary infections, thymoma, hypogammaglobulinemia, and absence of B cells. The median age of GS diagnosis was 57 years. During follow-up, immunoglobin replacement therapy effectively minimized sinopulmonary infections. However, the majority experienced severe and systemic viral or fungal infections, 3 developed basal cell carcinomas, and 5 had progressive bronchiectasis and persistent splenomegaly. The most notable clinical feature was opportunistic infections and in vitro evidence of cellular immune deficiency, which resulted in the death of 2 individuals. We also report a statistically significant, multidecade progressive decline in lymphocytes, platelets, hemoglobin, and red blood cells in our cohort, suggesting gradual bone marrow failure. CONCLUSIONS: Knowledge of the unique phenotype and temporal evolution of GS has allowed us to develop a more comprehensive diagnostic framework. It can be investigated as part of broader research into disease pathophysiology.


Subject(s)
Agammaglobulinemia , Immunologic Deficiency Syndromes , Primary Immunodeficiency Diseases , Thymoma , Thymus Neoplasms , Adult , Humans , Child , Middle Aged , Prospective Studies , Cross-Sectional Studies , Longitudinal Studies , Thymus Neoplasms/diagnosis , Thymus Neoplasms/pathology , Thymoma/diagnosis , Thymoma/pathology , Agammaglobulinemia/diagnosis , Agammaglobulinemia/therapy , Primary Immunodeficiency Diseases/diagnosis
9.
Front Immunol ; 14: 1187959, 2023.
Article in English | MEDLINE | ID: mdl-37435083

ABSTRACT

Hemophagocytic inflammatory syndrome (HIS) is a rare form of secondary hemophagocytic lymphohistiocytosis caused by an impaired equilibrium between natural killer and cytotoxic T-cell activity, evolving in hypercytokinemia and multiorgan failure. In the context of inborn errors of immunity, HIS occurrence has been reported in severe combined immunodeficiency (SCID) patients, including two cases of adenosine deaminase deficient-SCID (ADA-SCID). Here we describe two additional pediatric cases of ADA-SCID patients who developed HIS. In the first case, HIS was triggered by infectious complications while the patient was on enzyme replacement therapy; the patient was treated with high-dose corticosteroids and intravenous immunoglobulins with HIS remission. However, the patient required HLA-identical sibling donor hematopoietic stem cell transplantation (HSCT) for a definitive cure of ADA-SCID, without HIS relapse up to 13 years after HSCT. The second patient presented HIS 2 years after hematopoietic stem cell gene therapy (GT), secondarily to Varicella-Zoster vaccination and despite CD4+ and CD8+ lymphocytes' reconstitution in line with other ADA SCID patients treated with GT. The child responded to trilinear immunosuppressive therapy (corticosteroids, Cyclosporine A, Anakinra). We observed the persistence of gene-corrected cells up to 5 years post-GT, without HIS relapse. These new cases of children with HIS, together with those reported in the literature, support the hypothesis that a major dysregulation in the immune system can occur in ADA-SCID patients. Our cases show that early identification of the disease is imperative and that a variable degree of immunosuppression could be an effective treatment while allogeneic HSCT is required only in cases of refractoriness. A deeper knowledge of immunologic patterns contributing to HIS pathogenesis in ADA-SCID patients is desirable, to identify new targeted treatments and ensure patients' long-term recovery.


Subject(s)
Agammaglobulinemia , Lymphohistiocytosis, Hemophagocytic , Severe Combined Immunodeficiency , Humans , Child , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/therapy , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/therapy , Agammaglobulinemia/therapy , Cyclosporine
10.
J Clin Immunol ; 43(8): 1827-1839, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37454339

ABSTRACT

PURPOSE: X-linked agammaglobulinemia (XLA) is an inborn error of immunity caused by variants in Bruton's tyrosine kinase (BTK). XLA patients require lifelong immunoglobulin replacement therapy (IgRT). Only few XLA patients are indicated for allogeneic hematopoietic cell transplantation (HCT) because of severe complications. Accordingly, the published transplantation experience in XLA is minimal. We aimed to collect clinical data of XLA patients who received HCT in an international framework and to establish appropriate transplantation criteria and methods for XLA patients. METHODS: XLA patients were recruited through a questionnaire and a literature review. The data are on patient characteristics and transplantation methods and outcomes. RESULTS: In this study, twenty-two XLA patients who underwent HCT were recruited. The indication for HCT was recurrent or life-threatening infection in sixteen patients, malignancy in three, and other factors in three. A myeloablative conditioning, reduced toxicity myeloablative conditioning (RT-MAC), and reduced intensity conditioning (RIC) were selected in four, ten, and eight patients, respectively. Engraftment was achieved in 21 patients (95%). In all patients, 2-year overall survival (OS) and event-free survival (EFS) were 86% and 77%, respectively. In patients who received RT-MAC or RIC using treosulfan, busulfan, or melphalan, 2-year OS and EFS were 82% and 71%, respectively. Finally, twenty-one patients (95%) obtained complete or stable high-level mixed chimerism (50-95%), and the 1-year discontinuation rate of IgRT was 89%. CONCLUSION: Based on the concept in which IgRT is the standard treatment for XLA, HCT may be an effective and safe alternative treatment option for XLA patients, and IgRT can be discontinued following transplantation. It is ideal to perform HCT in XLA patients for whom transplantation is indicated before they develop organ damage.


Subject(s)
Agammaglobulinemia , Genetic Diseases, X-Linked , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Hematopoietic Stem Cell Transplantation/methods , Agammaglobulinemia/diagnosis , Agammaglobulinemia/therapy , Agammaglobulinemia/etiology , Genetic Diseases, X-Linked/therapy , Genetic Diseases, X-Linked/etiology , Melphalan , Transplantation Conditioning/methods , Graft vs Host Disease/etiology
11.
J Clin Immunol ; 43(7): 1623-1639, 2023 10.
Article in English | MEDLINE | ID: mdl-37328647

ABSTRACT

Purine nucleoside phosphorylase deficient severe combined immunodeficiency (PNP SCID) is one of the rare autosomal recessive primary immunodeficiency disease, and the data on epidemiology and outcome are limited. We report the successful management of a child with PNP SCID and present a systematic literature review of published case reports, case series, and cohort studies on PNP SCID listed in PubMed, Web of Science, and Scopus from 1975 until March 2022. Forty-one articles were included from the 2432 articles retrieved and included 100 PNP SCID patients worldwide. Most patients presented with recurrent infections, hypogammaglobulinaemia, autoimmune manifestations, and neurological deficits. There were six reported cases of associated malignancies, mainly lymphomas. Twenty-two patients had undergone allogeneic hematopoietic stem cell transplantation with full donor chimerism seen mainly in those receiving matched sibling donors and/or conditioning chemotherapy before the transplant. This research provides a contemporary, comprehensive overview on clinical manifestations, epidemiology, genotype mutations, and transplant outcome of PNP SCID. These data highlight the importance of screening for PNP SCID in cases presented with recurrent infections, hypogammaglobulinaemia, and neurological deficits.


Subject(s)
Agammaglobulinemia , Severe Combined Immunodeficiency , Child , Humans , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Purine-Nucleoside Phosphorylase/genetics , Agammaglobulinemia/diagnosis , Agammaglobulinemia/therapy , Agammaglobulinemia/complications , Reinfection/complications , Mutation
12.
Clin Lymphoma Myeloma Leuk ; 23(10): 719-732, 2023 10.
Article in English | MEDLINE | ID: mdl-37353432

ABSTRACT

Secondary antibody deficiency (SAD) is a subtype of secondary immunodeficiency characterized by low serum antibody concentrations (hypogammaglobulinemia) or poor antibody function. SAD is common in patients with multiple myeloma (MM) due to underlying disease pathophysiology and treatment-related immune system effects. Patients with SAD are more susceptible to infections and infection-related morbidity and mortality. With therapeutic advancements improving MM disease control and survival, it is increasingly important to recognize and treat the often-overlooked concurrent immunodeficiency present in patients with MM. The aims of this review are to define SAD and its consequences in MM, increase SAD awareness, and provide recommendations for SAD management. Based on expert panel discussions at a standalone meeting and supportive literature, several recommendations were made. Firstly, all patients with MM should be suspected to have SAD regardless of serum antibody concentrations. Patients should be evaluated for immunodeficiency at MM diagnosis and stratified into management categories based on their individualized risk of SAD and infection. Infection-prevention strategy education, early infection reporting, and anti-infective prophylaxis are key. We recommend prophylactic antibiotics or immunoglobulin replacement therapy (IgRT) should be considered in patients with severe hypogammaglobulinemia associated with a recurrent or persistent infection. To ensure an individualized and efficient treatment approach is utilized, patient's immunoglobin G concentration and infection burden should be closely monitored throughout treatment. Patient choice regarding route and IgRT treatment is also key in reducing treatment burden. Together, these recommendations and proposed management algorithms can be used to aid physician decision-making to improve patient outcomes.


Subject(s)
Agammaglobulinemia , Immunologic Deficiency Syndromes , Multiple Myeloma , Humans , Multiple Myeloma/complications , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Agammaglobulinemia/therapy , Immunologic Deficiency Syndromes/drug therapy , Immunization, Passive , Antibodies/therapeutic use
13.
Int J Exp Pathol ; 104(4): 154-176, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37177842

ABSTRACT

Rare diseases collectively exact a high toll on society due to their sheer number and overall prevalence. Their heterogeneity, diversity, and nature pose daunting clinical challenges for both management and treatment. In this review, we discuss recent advances in clinical applications of gene therapy for rare diseases, focusing on a variety of viral and non-viral strategies. The use of adeno-associated virus (AAV) vectors is discussed in the context of Luxturna, licenced for the treatment of RPE65 deficiency in the retinal epithelium. Imlygic, a herpes virus vector licenced for the treatment of refractory metastatic melanoma, will be an example of oncolytic vectors developed against rare cancers. Yescarta and Kymriah will showcase the use of retrovirus and lentivirus vectors in the autologous ex vivo production of chimeric antigen receptor T cells (CAR-T), licenced for the treatment of refractory leukaemias and lymphomas. Similar retroviral and lentiviral technology can be applied to autologous haematopoietic stem cells, exemplified by Strimvelis and Zynteglo, licenced treatments for adenosine deaminase-severe combined immunodeficiency (ADA-SCID) and ß-thalassaemia respectively. Antisense oligonucleotide technologies will be highlighted through Onpattro and Tegsedi, RNA interference drugs licenced for familial transthyretin (TTR) amyloidosis, and Spinraza, a splice-switching treatment for spinal muscular atrophy (SMA). An initial comparison of the effectiveness of AAV and oligonucleotide therapies in SMA is possible with Zolgensma, an AAV serotype 9 vector, and Spinraza. Through these examples of marketed gene therapies and gene cell therapies, we will discuss the expanding applications of such novel technologies to previously intractable rare diseases.


Subject(s)
Agammaglobulinemia , Severe Combined Immunodeficiency , Humans , Rare Diseases/genetics , Rare Diseases/therapy , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Genetic Therapy , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy
14.
J Clin Immunol ; 43(6): 1468-1477, 2023 08.
Article in English | MEDLINE | ID: mdl-37219739

ABSTRACT

PURPOSE: To understand the natural history and clinical outcomes for patients with X-linked agammaglobulinemia (XLA) in the United States utilizing the United States Immunodeficiency Network (USIDNET) patient registry. METHODS: The USIDNET registry was queried for data from XLA patients collected from 1981 to 2019. Data fields included demographics, clinical features before and after diagnosis of XLA, family history, genetic mutation in Bruton's tyrosine kinase (BTK), laboratory findings, treatment modalities, and mortality. RESULTS: Data compiled through the USIDNET registry on 240 patients were analyzed. Patient year of birth ranged from 1945 to 2017. Living status was available for 178 patients; 158/178 (88.8%) were alive. Race was reported for 204 patients as follows: White, 148 (72.5%); Black/African American, 23 (11.2%); Hispanic, 20 (9.8%); Asian or Pacific Islander, 6 (2.9%), and other or more than one race, 7 (3.4%). The median age at last entry, age at disease onset, age at diagnosis, and length of time with XLA diagnosis was 15 [range (r) = 1-52 years], 0.8 [r = birth-22.3 years], 2 [r = birth-29 years], and 10 [r = 1-56 years] years respectively. One hundred and forty-one patients (58.7%) were < 18 years of age. Two hundred and twenty-one (92%) patients were receiving IgG replacement (IgGR), 58 (24%) were on prophylactic antibiotics, and 19 (7.9%) were on immunomodulatory drugs. Eighty-six (35.9%) patients had undergone surgical procedures, two had undergone hematopoietic cell transplantation, and two required liver transplantation. The respiratory tract was the most affected organ system (51.2% of patients) followed by gastrointestinal (40%), neurological (35.4%), and musculoskeletal (28.3%). Infections were common both before and after diagnosis, despite IgGR therapy. Bacteremia/sepsis and meningitis were reported more frequently before XLA diagnosis while encephalitis was more commonly reported after diagnosis. Twenty patients had died (11.2%). The median age of death was 21 years (range = 3-56.7 years). Neurologic condition was the most common underlying co-morbidity for those XLA patients who died. CONCLUSIONS: Current therapies for XLA patients reduce early mortality, but patients continue to experience complications that impact organ function. With improved life expectancy, more efforts will be required to improve post-diagnosis organ dysfunction and quality of life. Neurologic manifestations are an important co-morbidity associated with mortality and not yet clearly fully understood.


Subject(s)
Agammaglobulinemia , Genetic Diseases, X-Linked , Humans , Infant , Child, Preschool , Child , Adolescent , Young Adult , Adult , Middle Aged , Quality of Life , Agammaglobulinaemia Tyrosine Kinase/genetics , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/epidemiology , Genetic Diseases, X-Linked/genetics , Agammaglobulinemia/diagnosis , Agammaglobulinemia/epidemiology , Agammaglobulinemia/therapy , Mutation/genetics
15.
Rev Med Suisse ; 19(821): 668-673, 2023 Apr 05.
Article in French | MEDLINE | ID: mdl-37017348

ABSTRACT

Infections are among the leading causes of morbidity and mortality in lymphoproliferative malignancies such as multiple myeloma (MM) and chronic lymphocytic leukemia (CLL). The causes of infections are often multifactorial and may be due to disease- or treatment-related factors. New therapies have improved survival in lymphoproliferative malignancies, resulting in an increased incidence of secondary immune deficiencies (SID) in these diseases.


Les infections sont l'une des principales causes de morbidité et de mortalité dans les maladies lymphoprolifératives malignes telles que le myélome multiple (MM) et la leucémie lymphoïde chronique (LLC). Les causes des infections sont souvent multifactorielles et peuvent être secondaires à des facteurs liés à la maladie ou au traitement. Les nouvelles thérapies ont permis d'améliorer le taux de survie des maladies lymphoprolifératives malignes, ce qui a entraîné une augmentation de l'incidence des déficits immunitaires secondaires (DIS) dans ces maladies.


Subject(s)
Agammaglobulinemia , Immunologic Deficiency Syndromes , Leukemia, Lymphocytic, Chronic, B-Cell , Multiple Myeloma , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/complications , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Multiple Myeloma/complications , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Agammaglobulinemia/complications , Agammaglobulinemia/therapy , Immunologic Deficiency Syndromes/complications
17.
J Allergy Clin Immunol Pract ; 11(6): 1665-1675, 2023 06.
Article in English | MEDLINE | ID: mdl-36736952

ABSTRACT

Inherited defects in the adenosine deaminase (ADA) gene typically cause severe combined immunodeficiency. In addition to infections, ADA-deficient patients can present with neurodevelopmental, behavioral, hearing, skeletal, lung, heart, skin, kidney, urogenital, and liver abnormalities. Some patients also suffer from autoimmunity and malignancies. In recent years, there have been remarkable advances in the management of ADA deficiency. Most ADA-deficient patients can be identified by newborn screening for severe combined immunodeficiency, which facilitates early diagnosis and treatment of asymptomatic infants. Most patients benefit from enzyme replacement therapy (ERT). Allogeneic hematopoietic cell transplantation from an HLA-matched sibling donor or HLA-matched family member donor with no conditioning is currently the preferable treatment. When matched sibling donor or matched family member donor is not available, autologous ADA gene therapy with nonmyeloablative conditioning and ERT withdrawal, which is reported in recent studies to result in 100% overall survival and 90% to 95% engraftment, should be pursued. If gene therapy is not immediately available, ERT can be continued for a few years, although its excessive cost might be prohibitive. The recent improved outcome of hematopoietic cell transplantation using HLA-mismatched family-related donors or HLA-matched unrelated donors, after reduced-intensity conditioning, suggests that such procedures might also be considered rather than continuing ERT for prolonged periods. Long-term follow-up will further assist in determining the optimal treatment approach for ADA-deficient patients.


Subject(s)
Agammaglobulinemia , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Humans , Infant , Infant, Newborn , Adenosine Deaminase/genetics , Agammaglobulinemia/diagnosis , Agammaglobulinemia/therapy , Agammaglobulinemia/genetics , Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy
18.
Kyobu Geka ; 76(2): 115-118, 2023 Feb.
Article in Japanese | MEDLINE | ID: mdl-36731844

ABSTRACT

Thymoma presenting concurrent pure red-cell aplasia (PRCA) and hypogammaglobulinemia are extremely rare. A 67-year-old woman with a short of breath was referred to our hospital due to anemia and the chest abnormal shadow. Laboratory investigations revealed a hemoglobin level of 5.6 g/dl and reticulocyte percentage of 0.2%. Her serum gamma-globulin level was low. Chest computed tomography (CT) revealed a 7-cm tumor in the left upper mediastinum. We diagnosed the patient with thymoma accompanied by PRCA and hypogammaglobulinemia. The patient underwent thymectomy and PRCA has been successfully treated by postoperative cyclosporine administration. Monthly intravenous infusion of gamma-globulin has been necessary for the control of hypogammaglobulinemia. Currently, she is doing well without recurrence of thymoma or PRCA five years after the surgery.


Subject(s)
Agammaglobulinemia , Red-Cell Aplasia, Pure , Thymoma , Thymus Neoplasms , Humans , Female , Aged , Thymoma/complications , Thymoma/diagnostic imaging , Thymoma/surgery , Agammaglobulinemia/complications , Agammaglobulinemia/therapy , Thymus Neoplasms/complications , Thymus Neoplasms/diagnostic imaging , Thymus Neoplasms/surgery , Red-Cell Aplasia, Pure/complications , Red-Cell Aplasia, Pure/pathology , Thymectomy , gamma-Globulins
19.
Rev Med Interne ; 44(3): 133-138, 2023 Mar.
Article in French | MEDLINE | ID: mdl-36725480

ABSTRACT

Hypogammaglobulinemia (hypoγ) is defined as a serum IgG level < 7 g/L. It is most often detected on serum protein electrophoresis. Given the existence of transient hypoγ, its persistence should be checked at distance, preferably by requesting a blood test for IgG, IgA and IgM, which will be needed to characterize a possible primary immune deficiency (PID). In the case of association with a monoclonal component, the first step is to look for a cryoglobulin causing a false hypoγ. Otherwise, the etiological investigation is dictated by the clinical examination. For example, the notion of chronic diarrhea should lead to a search for an enteropathy causing a digestive loss of gammaglobulins (an ambiguous situation because some DIP can be complicated by an enteropathy). In the absence of an obvious explanation, a secondary cause must first be ruled out (secondary immune deficiencies are 30 times more common than PID). The first simple test to perform is 24-hour proteinuria, coupled with urinary protein electrophoresis, to rule out 2 diagnoses: nephrotic syndrome and light chain myeloma. Subsequently, blood immunophenotyping looking for a circulating B clone is recommended, allowing the investigations to be directed towards a lymphoid hemopathy. Drug-induced hypoγ may also be suspected if certain drugs such as corticosteroids, anti-epileptics or immunosuppressive agents (especially anti-CD20) are taken. The profile of a drug-induced hypoγ is different from that of a DIP: it is rarely profound, the IgA level is preserved and there is no deficit in switched memory B lymphocytes. Finally, a thoracoabdominal CT-scan will help to rule out a thymoma and identify a deep tumor syndrome. If all these tests are normal, a PID is suspected, the leader of which in adults remains the common variable immunodeficiency, which is the most frequent symptomatic PID in adults.


Subject(s)
Agammaglobulinemia , Common Variable Immunodeficiency , Thymus Neoplasms , Adult , Humans , Agammaglobulinemia/complications , Agammaglobulinemia/diagnosis , Agammaglobulinemia/therapy , Thymus Neoplasms/complications , Immunoglobulin A , Immunoglobulin G
20.
J Pediatr Hematol Oncol ; 45(1): e145-e149, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36598967

ABSTRACT

Rituximab (RTX) is widely employed to treat Epstein-Barr virus reactivation in children undergoing Hematopoietic Cell Transplantation (HCT). The resulting loss of B cells may cause persistent hypogammaglobulinemia. This retrospective cross-sectional study aims to identify flow cytometry biomarkers associated with persistent hypogammaglobulinemia in patients receiving RTX after HCT. We analyzed 5 patients (cases group) requiring immunoglobulin substitution due to low level of IgG (IgG <5 g/L) detected after RTX treatment and 5 patients (controls group) not requiring long-term immunoglobulin (Ig) substitution. We investigated the B cell reconstitution, and in patients group we observed a significantly lower count in B total, IgD+CD27+ marginal B cells and IgD-CD27+ switched-memory B cells, after a median of 5 years from HCT, compared with the control group. Despite the importance limits of our study and the heterogeneity of our data (age of included patients, time of evaluation, interval between RTX dose and assessment) we conclude that RTX given early after HCT might cause a deranged B cell maturation, contributing to the delation in B cell recovery following HCT, and switched memory and marginal zone B cell counts could be a promising biomarker to identify patients requiring long-term Ig substitution.


Subject(s)
Agammaglobulinemia , B-Lymphocyte Subsets , Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Humans , Child , Rituximab/therapeutic use , Agammaglobulinemia/therapy , Agammaglobulinemia/chemically induced , Retrospective Studies , Cross-Sectional Studies , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/etiology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Herpesvirus 4, Human , Hematopoietic Stem Cell Transplantation/adverse effects , Biomarkers , Immunoglobulin G
SELECTION OF CITATIONS
SEARCH DETAIL
...