ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Severe combined immunodeficiency (SCID): Specific defects

Severe combined immunodeficiency (SCID): Specific defects
Literature review current through: Jan 2024.
This topic last updated: Jan 30, 2024.

INTRODUCTION — Combined immunodeficiency syndromes encompass a rapidly expanding heterogeneous group of both clinically and genetically defined disorders arising from a disturbance in the development and function of both T cell and B cell (cellular and humoral) arms of the adaptive immune system (figure 1 and table 1). These disorders are termed "severe" (eg, severe combined immune deficiency [SCID]) when there is a complete absence of T cell function, which leads to early death from overwhelming infection, typically in the first year of life, without definitive treatment [1].

Brief synopses are given for the molecular types of SCID that are discussed in detail separately:

X-linked SCID (see "X-linked severe combined immunodeficiency (X-SCID)")

Adenosine deaminase (ADA) deficiency (see "Adenosine deaminase deficiency: Pathogenesis, clinical manifestations, and diagnosis")

Artemis, recombination-activating gene 1 (RAG1), recombination-activating gene 2 (RAG2), and deoxyribonucleic acid protein kinase catalytic subunit (DNA-PKcs) deficiencies (see "T-B-NK+ SCID: Pathogenesis, clinical manifestations, and diagnosis" and "T-B-NK+ SCID: Management")

Janus kinase 3 (JAK3) deficiency (see "Severe combined immunodeficiency (SCID) with JAK3 deficiency")

Deficiencies in CD3 complex components (see "CD3/T cell receptor complex disorders causing immunodeficiency")

A general overview of SCID is also presented separately. (See "Severe combined immunodeficiency (SCID): An overview".)

Combined immunodeficiencies that are not considered "severe" are discussed separately:

(See "Combined immunodeficiencies: An overview" and "Combined immunodeficiencies: Specific defects".)

(See "CD3/T cell receptor complex disorders causing immunodeficiency".)

(See "Purine nucleoside phosphorylase deficiency".)

(See "ZAP-70 deficiency".)

CATEGORIZATION BASED UPON GENOTYPES AND PHENOTYPES — Pathogenic variants of a particular gene may either lead to SCID or to a milder immunodeficiency, depending upon whether the defect is complete or partial. Gene defects that lead to partial function of the gene product are called "hypomorphic," whereas complete defects are called "null" or "amorphic." Based upon the 2022 International Union of Immunology Societies (IUIS) report, there are 18 genetically defined forms of SCID that are generally categorized based upon the presence or absence of B and/or natural killer (NK) cells in addition to an absence of T cells [2]. The Primary Immune Deficiency Treatment Consortium (PIDTC) published updated diagnostic guidelines for SCID in 2023 [3].

Although the sections below group SCID genotypes by their classical phenotypic presentation as it relates to populations of T, B, and NK cells, there is an increasing awareness of phenotypic variation within a genotype. Thus, broad genetic testing is critical to determine which genetic form of SCID a patient has, rather than either relying on lymphocyte phenotyping or narrowly focused genetic testing. Awareness of the genotype has implications in the immediate supportive therapy, considerations for approach to definitive therapy, risk of late effects after definitive therapy, and implications for later family planning/genetic counseling.

T-B+NK- SCID — Gene pathogenic variants affecting the integrity of the gamma-c/Janus kinase 3 (JAK3) signaling pathway result in T cell-negative, B cell-positive, natural killer-negative (T-B+NK-) SCID and include X-linked SCID and JAK3 deficiency.

X-linked SCID — X-linked SCID (MIM #300400), the most common form of typical SCID, is due to defects in interleukin (IL) 2 receptor subunit gamma (IL2RG), which encodes the common gamma chain of the IL-2 receptor shared by five other cytokine receptors (IL-4, 7, 9, 15, and 21). These male patients usually have the classic clinical SCID phenotype, presenting in the newborn period with recurrent severe infections, chronic diarrhea, and failure to thrive if they are not diagnosed via newborn screening (NBS) for SCID. Gene therapy has been developed and used in clinical trials with some early success for this form of SCID. However, late development of leukemia in some patients receiving gamma retroviral-based therapy led to a need to develop safer vectors, which are available only on a research basis [4]. This form of SCID is discussed in greater detail separately. (See "X-linked severe combined immunodeficiency (X-SCID)".)

JAK3 deficiency — Janus kinase 3 (JAK3; encoded on chromosome 19p12-13.1) mediates cytokine signal transduction via gamma-c. This autosomal recessive form of T-B+NK- SCID (MIM #600802) is identical to X-linked SCID in cellular and clinical phenotypes. Rarely, partial JAK3 defects associated with low amounts of functional protein can present with mild immunodeficiency. SCID due to JAK3 deficiency is discussed in greater detail separately. (See "Severe combined immunodeficiency (SCID) with JAK3 deficiency".)

T-B+NK+ SCID — The T cell-negative, B cell-positive, natural killer-positive (T-B+NK+) SCID syndromes include defects in IL-7 receptor alpha chain (IL7RA; also called CD127); CD45 (also called protein-tyrosine phosphatase, receptor-type, C [PTPRC]); the CD3 chains: CD3 delta (CD3D), CD3 epsilon (CD3E), and CD3 zeta (CD3Z); linker for activation of T cells (LAT); and coronin 1A (CORO1A).

IL-7 receptor alpha chain (CD127) deficiency — The IL7RA chain gene (IL7R, encoded on chromosome 5p13) plays a critical role in cytokine signaling that is necessary for T cell development. Deficiency of the IL7RA chain (MIM #608971) is among the most common types of SCID, and these patients have a typical SCID phenotype [5-7]. Pathogenic variants in the IL7R gene can also present with an Omenn syndrome phenotype [8]. (See "T-B-NK+ SCID: Pathogenesis, clinical manifestations, and diagnosis", section on 'Omenn syndrome phenotype' and "T-B-NK+ SCID: Pathogenesis, clinical manifestations, and diagnosis", section on 'T-B-NK+ SCID without radiation sensitivity due to RAG defects (includes most cases of Omenn syndrome)'.)

CD45 deficiency — CD45, the leukocyte common antigen (also called protein-tyrosine phosphatase, receptor-type, C [PTPRC]), is encoded on chromosome 1q31-q32. CD45 is a transmembrane tyrosine phosphatase involved in T cell receptor (TCR) signaling and T cell development in the thymus. Only a few patients with classic SCID due to CD45 deficiency (MIM #608971) have been described [9-11].

CD3 complex component deficiencies — The CD3 complex plays a major role in signaling through the TCR. Pathogenic variants in the genes encoding CD3 chains (CD3D, CD3E, and CD3Z) lead to SCID (MIM #615617, #615615, #610163). Patients with CD3 gamma deficiency have varying phenotypes, with some having SCID-like symptoms and some having a more benign course, often marked by immune dysregulation. CD3 chain deficiencies are discussed in greater detail separately. (See "CD3/T cell receptor complex disorders causing immunodeficiency", section on 'CD3 deficiency'.)

Actin-regulating protein coronin 1A deficiency — CORO1A (encoded on chromosome 16p11.2) is involved in actin cytoskeleton regulation and is essential for T cell egress from the thymus. Autosomal recessive defects in CORO1A lead to absence of normal peripheral T cells and a typical T-B+NK+ SCID phenotype (MIM #615401) with a visible thymus on chest radiograph [12-17]. Other reported features include Epstein-Barr virus-induced B cell lymphoproliferative syndrome and B cell lymphoma at an early age and attention deficit hyperactivity disorder.

LAT deficiency — Autosomal recessive pathogenic variants in the linker for activation of T cells (LAT) gene lead to a T-B+NK+ SCID phenotype (MIM #617514). The LAT protein plays a crucial role in the process of linking TCR activation to downstream intracellular T cell responses. In addition to immunodeficiency, affected patients also experience severe autoimmune disease, particularly cytopenias. (See "CD3/T cell receptor complex disorders causing immunodeficiency", section on 'LAT deficiency'.)

T-B-NK+ SCID — Genetic defects that disrupt both T and B cell development with preservation of natural killer (NK) cells result in T cell-negative, B cell-negative, NK cell-positive SCID (T-B+NK+ SCID). These SCID syndromes include the V(D)J recombination defects due to autosomal recessive defects in recombination-activating genes 1 and 2 (RAG1 and RAG2). However, the other genotypes within this phenotypic category have SCID associated with radiosensitivity. These include DNA cross-link repair protein 1C (DCLRE1C, the gene for Artemis); protein kinase, DNA-activated, catalytic subunit (PRKDC, also called DNA protein kinase catalytic subunit [DNA-PKcs]); nonhomologous end-joining factor 1 (NHEJ1, also called Cernunnos or XRCC4-like factor [XLF]); and DNA ligase IV (LIG4). Patients with radiosensitive forms of SCID are at higher risk of poor survival, complications from exposure to alkylating agents during predefinitive therapy conditioning, and higher risk of late medical complications associated both with therapy and the underlying genetic disease [18-20]. These defects are reviewed briefly here and discussed in greater detail separately. (See "T-B-NK+ SCID: Pathogenesis, clinical manifestations, and diagnosis" and "T-B-NK+ SCID: Management".)

RAG1/RAG2 deficiency — Recombination-activating gene 1 (RAG1) and recombination-activating gene 2 (RAG2) are expressed exclusively in lymphocytes and mediate the creation of double-strand DNA breaks at the sites of recombination and signal sequences during T and B cells receptor gene rearrangement. There has been an increased detection of SCID caused by RAG1/RAG2 defects (MIM #601457), particularly leaky forms of SCID, since the widespread use of SCID newborn screening (NBS) was implemented in the United States [1]. Nonhomologous end joining is normal in individuals with RAG1 or RAG2 deficiency. Autosomal recessive pathogenic variants in RAG1 and RAG2 are not associated with radiation sensitivity, which has implications for the types of conditioning used prior to hematopoietic cell transplantation for these patients.

DCLRE1C (Artemis) deficiency — Artemis deficiency (MIM #602452) is also known as Athabascan SCID (SCIDA) since a founder pathogenic variant in DNA cross-link repair 1C (DCLRE1C) is found with increased frequency in Native Americans speaking one of the Athabascan family languages (eg, Apache, Navajo). Pathogenic variants in DCLRE1C lead to T-B-NK+ SCID because the Artemis protein is critical to the end-processing step of nonhomologous end joining necessary for V(D)J recombination in T and B cell receptor development. Patients with this form of SCID also demonstrate increased sensitivity to ionizing radiation and alkylator-based chemotherapy used in many preparative regimens for hematopoietic cell transplantation since Artemis is also required for repair of double-strand DNA breaks caused by these insults. A clinical trial of gene therapy for Artemis SCID is available in the US [4].

DNA protein kinase catalytic subunit (DNA-PKcs) deficiency — DNA-PKcs has a role in rejoining double-strand breaks in DNA critical to nonhomologous end joining necessary for V(D)J recombination in T and B cell receptor development. Autosomal recessive pathogenic variants in PRKDC, the gene for DNA-PKcs, lead to an inability to develop T and B cells (MIM #615966). These patients also demonstrate radiosensitivity and sensitivity to alkylator-based conditioning regimens, similar to patients with SCIDA.

Cernunnos/XLF deficiency — Nonhomologous end-joining factor 1 protein (NHEJ1, also called Cernunnos or XRCC4-like factor [XLF]) is involved in the end-bridging and ligation steps of nonhomologous end joining necessary for V(D)J recombination in T and B cell receptor development. Autosomal recessive pathogenic variants in NHEJ1 cause Cernunnos/XLF deficiency (MIM #611291). In addition to T-B-NK+ SCID, patients with this genetic defect demonstrate microcephaly and poor growth. These patients also demonstrate radiosensitivity and sensitivity to alkylator-based conditioning regimens, similar to patients with SCIDA.

DNA ligase IV deficiency — DNA ligase IV plays a role in rejoining double-strand breaks in DNA critical to nonhomologous end joining necessary for V(D)J recombination in T and B cell receptor development. Autosomal recessive pathogenic variants in LIG4, the gene for DNA ligase IV, lead to an inability to develop T and B cells (MIM #611291). Other commonly associated features are microcephaly and growth failure, similar to patients with Cernunnos/XLF deficiency, and radiosensitivity and sensitivity to alkylator-based conditioning regimens, similar to patients with SCIDA.

Activated RAC2 defect — Ras-related C3 botulinum toxin substrate 2 (RAC2) has a role in neutrophil superoxide production as well actin remodeling. Autosomally dominant inherited gain-of-function pathogenic variants in RAC2 have led to T and B cell lymphopenia as well as excessive neutrophil superoxide production in patients (MIM #618986) [21-24]. Some patients were diagnosed following abnormal T cell receptor excision circle (TREC) based NBS for SCID, while others presented with clinical symptoms of recurrent bacterial and viral infections, lymphoproliferation, and neutropenia.

T-B-NK- SCID — T cell-negative, B cell-negative, natural killer cell-negative (T-B-NK-) SCID syndromes include adenosine deaminase (ADA) deficiency and reticular dysgenesis.

Adenosine deaminase deficiency — ADA deficiency (MIM #102700) accounts for approximately 10 to 15 percent of SCID. Approximately 90 percent of patients with ADA deficiency have a classic severe SCID phenotype, and they develop severe infections in the first months of life. Most of the remainder have a "delayed" or "late-onset" form that presents in late infancy or early childhood. Definitive treatments included hematopoietic cell transplantation and gene therapy. Polyethylene glycol (PEG) ADA is a reasonably effective therapeutic agent to use as a bridge to definitive therapy. (See "Adenosine deaminase deficiency: Pathogenesis, clinical manifestations, and diagnosis" and "Adenosine deaminase deficiency: Treatment and prognosis" and "Overview of gene therapy for inborn errors of immunity", section on 'Adenosine deaminase deficiency SCID'.)

Reticular dysgenesis — Reticular dysgenesis (MIM #267500) is one of the rarest and most severe forms of SCID [25-36]. Infants with reticular dysgenesis are often born prematurely and are small for gestational age. Severe infections occur earlier than in other forms of SCID due to profound neutropenia in addition to markedly decreased T and NK cells and absent to low normal B cells. Neutrophil counts do not increase with granulocyte colony-stimulating factor (G-CSF) treatment. Other features include bilateral sensorineural deafness and skeletal abnormalities (squaring of scapular tips and cupping and fraying of the anterior rib costochondral junctions). This autosomal recessive SCID is caused by pathogenic variants in the mitochondrial adenylate kinase 2 (AK2) gene.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Inborn errors of immunity (previously called primary immunodeficiencies)".)

SUMMARY

Terminology – Severe combined immunodeficiency (SCID) is an umbrella term for a group of clinical disorders that arise from pathogenic variants in genes whose products are essential for the normal development and function of T and B cells (cellular and humoral immunity) (figure 1 and table 1). In patients with a typical presentation, there is a complete absence of T cell-mediated immune function that typically leads to death from overwhelming infection in the first year or so of life if not treated with definitive therapy (hematopoietic cell therapy or gene therapy). (See 'Introduction' above.)

Classification – The classic approach has been to classify SCID syndromes as T-B+NK+, T-B+NK-, T-B-NK+, or T-B-NK- based upon the presence of defects affecting T cells and with or without defects also affecting B and/or natural killer (NK) cells (table 1). This paradigm is gradually being replaced by naming these conditions based upon their specific molecular causes. (See 'T-B+NK- SCID' above and 'T-B+NK+ SCID' above and 'T-B-NK+ SCID' above and 'T-B-NK- SCID' above.)

Impact of genotype on treatment – Knowledge of the gene responsible for the expressed phenotype can have important implications for approach to and choice of definitive treatment, such as in the case of T-B-NK+, where the presence of radiosensitivity affects the choice of pretransplant conditioning regimen, or whether gene therapy is an alternative to hematopoietic cell transplantation. (See 'T-B-NK+ SCID' above and 'Adenosine deaminase deficiency' above and "Overview of gene therapy for inborn errors of immunity" and "Hematopoietic cell transplantation for severe combined immunodeficiencies".)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Francisco A Bonilla, MD, PhD, who contributed to earlier versions of this topic review.

  1. Kwan A, Abraham RS, Currier R, et al. Newborn screening for severe combined immunodeficiency in 11 screening programs in the United States. JAMA 2014; 312:729.
  2. Bousfiha A, Moundir A, Tangye SG, et al. The 2022 Update of IUIS Phenotypical Classification for Human Inborn Errors of Immunity. J Clin Immunol 2022; 42:1508.
  3. Dvorak CC, Haddad E, Heimall J, et al. The diagnosis of severe combined immunodeficiency (SCID): The Primary Immune Deficiency Treatment Consortium (PIDTC) 2022 Definitions. J Allergy Clin Immunol 2023; 151:539.
  4. Kohn LA, Kohn DB. Gene Therapies for Primary Immune Deficiencies. Front Immunol 2021; 12:648951.
  5. Roifman CM, Zhang J, Chitayat D, Sharfe N. A partial deficiency of interleukin-7R alpha is sufficient to abrogate T-cell development and cause severe combined immunodeficiency. Blood 2000; 96:2803.
  6. Puel A, Leonard WJ. Mutations in the gene for the IL-7 receptor result in T(-)B(+)NK(+) severe combined immunodeficiency disease. Curr Opin Immunol 2000; 12:468.
  7. Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T(-)B(+)NK(+) severe combined immunodeficiency. Nat Genet 1998; 20:394.
  8. Villa A, Notarangelo LD, Roifman CM. Omenn syndrome: inflammation in leaky severe combined immunodeficiency. J Allergy Clin Immunol 2008; 122:1082.
  9. Kung C, Pingel JT, Heikinheimo M, et al. Mutations in the tyrosine phosphatase CD45 gene in a child with severe combined immunodeficiency disease. Nat Med 2000; 6:343.
  10. Tchilian EZ, Wallace DL, Wells RS, et al. A deletion in the gene encoding the CD45 antigen in a patient with SCID. J Immunol 2001; 166:1308.
  11. Roberts JL, Buckley RH, Luo B, et al. CD45-deficient severe combined immunodeficiency caused by uniparental disomy. Proc Natl Acad Sci U S A 2012; 109:10456.
  12. Shiow LR, Roadcap DW, Paris K, et al. The actin regulator coronin 1A is mutant in a thymic egress-deficient mouse strain and in a patient with severe combined immunodeficiency. Nat Immunol 2008; 9:1307.
  13. Shiow LR, Paris K, Akana MC, et al. Severe combined immunodeficiency (SCID) and attention deficit hyperactivity disorder (ADHD) associated with a Coronin-1A mutation and a chromosome 16p11.2 deletion. Clin Immunol 2009; 131:24.
  14. Moshous D, Martin E, Carpentier W, et al. Whole-exome sequencing identifies Coronin-1A deficiency in 3 siblings with immunodeficiency and EBV-associated B-cell lymphoproliferation. J Allergy Clin Immunol 2013; 131:1594.
  15. Mace EM, Orange JS. Lytic immune synapse function requires filamentous actin deconstruction by Coronin 1A. Proc Natl Acad Sci U S A 2014; 111:6708.
  16. Stray-Pedersen A, Jouanguy E, Crequer A, et al. Compound heterozygous CORO1A mutations in siblings with a mucocutaneous-immunodeficiency syndrome of epidermodysplasia verruciformis-HPV, molluscum contagiosum and granulomatous tuberculoid leprosy. J Clin Immunol 2014; 34:871.
  17. Punwani D, Pelz B, Yu J, et al. Coronin-1A: immune deficiency in humans and mice. J Clin Immunol 2015; 35:100.
  18. Haddad E, Logan BR, Griffith LM, et al. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery. Blood 2018; 132:1737.
  19. Abd Hamid IJ, Slatter MA, McKendrick F, et al. Long-Term Health Outcome and Quality of Life Post-HSCT for IL7Rα-, Artemis-, RAG1- and RAG2-Deficient Severe Combined Immunodeficiency: a Single Center Report. J Clin Immunol 2018; 38:727.
  20. Eissa H, Thakar MS, Shah AJ, et al. Posttransplantation late complications increase over time for patients with SCID: A Primary Immune Deficiency Treatment Consortium (PIDTC) landmark study. J Allergy Clin Immunol 2024; 153:287.
  21. Hsu AP, Donkó A, Arrington ME, et al. Dominant activating RAC2 mutation with lymphopenia, immunodeficiency, and cytoskeletal defects. Blood 2019; 133:1977.
  22. Lougaris V, Chou J, Beano A, et al. A monoallelic activating mutation in RAC2 resulting in a combined immunodeficiency. J Allergy Clin Immunol 2019; 143:1649.
  23. Sharapova SO, Haapaniemi E, Sakovich IS, et al. Heterozygous activating mutation in RAC2 causes infantile-onset combined immunodeficiency with susceptibility to viral infections. Clin Immunol 2019; 205:1.
  24. Smits BM, Lelieveld PHC, Ververs FA, et al. A dominant activating RAC2 variant associated with immunodeficiency and pulmonary disease. Clin Immunol 2020; 212:108248.
  25. de VAAL O, SEYNHAEVE V. Reticular dysgenesia. Lancet 1959; 2:1123.
  26. GITLIN D, VAWTER G, CRAIG JM. THYMIC ALYMPHOPLASIA AND CONGENITAL ALEUKOCYTOSIS. Pediatrics 1964; 33:184.
  27. Ownby DR, Pizzo S, Blackmon L, et al. Severe combined immunodeficiency with leukopenia (reticular dysgenesis) in siblings: immunologic and histopathologic findings. J Pediatr 1976; 89:382.
  28. Español T, Compte J, Alvarez C, et al. Reticular dysgenesis: report of two brothers. Clin Exp Immunol 1979; 38:615.
  29. Levinsky RJ, Tiedeman K. Successful bone-marrow transplantation for reticular dysgenesis. Lancet 1983; 1:671.
  30. Small TN, Wall DA, Kurtzberg J, et al. Association of reticular dysgenesis (thymic alymphoplasia and congenital aleukocytosis) with bilateral sensorineural deafness. J Pediatr 1999; 135:387.
  31. Bertrand Y, Müller SM, Casanova JL, et al. Reticular dysgenesis: HLA non-identical bone marrow transplants in a series of 10 patients. Bone Marrow Transplant 2002; 29:759.
  32. Reubsaet LL, Boelens JJ, Rademaker C, et al. Successful cord blood transplantation in a premature and dysmature neonate of 1700 g with reticular dysgenesis. Bone Marrow Transplant 2007; 39:307.
  33. Pannicke U, Hönig M, Hess I, et al. Reticular dysgenesis (aleukocytosis) is caused by mutations in the gene encoding mitochondrial adenylate kinase 2. Nat Genet 2009; 41:101.
  34. Al-Zahrani D, Al-Ghonaium A, Al-Mousa H, et al. Skeletal abnormalities and successful hematopoietic stem cell transplantation in patients with reticular dysgenesis. J Allergy Clin Immunol 2013; 132:993.
  35. Hoenig M, Lagresle-Peyrou C, Pannicke U, et al. Reticular dysgenesis: international survey on clinical presentation, transplantation, and outcome. Blood 2017; 129:2928.
  36. Hoenig M, Pannicke U, Gaspar HB, Schwarz K. Recent advances in understanding the pathogenesis and management of reticular dysgenesis. Br J Haematol 2018; 180:644.
Topic 3912 Version 23.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟