Severe Combined Immunodeficiency (SCID)

Back

Practice Essentials

Severe combined immunodeficiency (SCID) is a group of medical disorders that result from genetic defects in both cellular and humoral immunity. Those immune defects lead to infections with bacterial, viral, and fungal pathogens that begin during infancy and, if untreated, result in a fatal outcome in the first few years of life.[1]  

There has been a considerable gain in knowledge of the pathologic conditions of the immune system since the recognition of primary immunodeficiency as an entity in 1950, highlighted by the discovery of X-linked agammaglobulinemia, congenital neutropenia, and SCID. Over 200 diseases with more than 300 genetic etiologies have been described, which has provided opportunities for diagnosis and genetic counseling.[2]  

Moreover, an understanding of the pathogenesis of primary immunodeficiencies has paved the way for immunologic interventions and new treatments, such as immunoglobulin G (IgG) replacement, bone marrow transplantation, and gene therapy. The discovery of the HLA system in 1968 led to successful bone marrow transplantations, and patients with immunodeficiency syndromes were the first to benefit from this novel therapy.

For patient education information, see the Immunodeficiency Disorders Directory.

Pathophysiology

B and T cells, type 2 dendritic cells, and natural killer (NK) cells share a common ancestor: the common lymphoid progenitor (CLP). CLP differentiates into 2 intermediate progenitors, termed early B cells and T/NK/DC trilineage cells. Both intermediate progenitors continue their development in the bone marrow through primary lymphopoiesis, which is an antigen-independent process. Secondary B-cell lymphopoiesis is an antigen-dependent process that occurs in the germinal centers of peripheral lymphoid organs with specific antibody production. Secondary T-cell lymphopoiesis is also antigen-dependent and occurs in the thymus.

The earlier the defect, the more devastating the effect on lymphopoiesis. Defects occurring at the CLP stage or those affecting processes common to B- and T-cell development result in SCID involving B, T, and NK cells. According to the type of defect that leads to a SCID phenotype, Combined B- and T-cell disorders can be divided into specific groups with unique pathophysiologies that invariably result in an absence of nonfunctional B cells and absence of T cells (see Table 1).

Table 1. Classification of SCID



View Table

See Table

Adapted from Cavazzana-Calvo M, Fischer A. Gene therapy for severe combined immunodeficiency: are we there yet? J Clin Invest. Jun 2007;117(6):1456-65.[3]

In other circumstances, the defect can affect later events in lymphopoiesis; a major loss or dysfunction in T cells can cause secondary B-cell deficiency, resulting in a clinical disorder that manifests as a combined B- and T-cell deficiency.

There are 4 characterized pathways that can result in SCID, as follows:

Defects in purine pathway enzymes that result in buildup of metabolites toxic to lymphocytes

ADA is an enzyme of the purine salvage pathway that is responsible for the deamination of adenosine to inosine and the deamination of deoxyadenosine to deoxyinosine. The deficiency of this enzyme leads to the accumulation of deoxyadenosine triphosphate (dATP) and 2'-deoxyadenosine. An increase in the intracellular levels of dATP is toxic to lymphocytes because it inhibits the enzyme ribonucleotide reductase, leading to suppression of DNA synthesis, whereas 2'-deoxyadenosine inhibits the enzyme S- adenosyl-L-homocysteine (SAH) hydrolase, which results in accumulation of SAH, a potent inhibitor of all cellular methylation reactions. Both B and T cells are affected, leading to SCID.

Defects in recombination of the antigen receptor genes (RAG) of B-cells and T-cells

Immunoglobulin gene rearrangement begins with heavy-chain gene rearrangement, which is followed by light-chain gene rearrangement. Once the rearrangement process is finished, recombination signal sequences that served to approximate the different genes from each other are removed with the help of the RAG1 and RAG2 proteins. RAG1/RAG2 deficiency is responsible for the B- and T-cell maturation defects in some persons with SCID.

Omenn syndrome is a rare, inherited disorder with a pooly understood pathogenesis. In this condition, mutations in the genes coding for the recombinases (ie, RAG1 and RAG2) cause a defect in the VDJ rearrangement that is needed for mature B-cells and T-cells to develop, resulting in a paradoxical combination of immunodeficiency and immune dysregulation.

In study by Khiong et al, the authors identified a C57BL/10 mouse with a spontaneous mutation in and reduced activity of RAG1.[4] Mice bred from this animal exhibited major manifestations of Omenn syndrome, including high numbers of memory-phenotype T cells, hepatosplenomegaly and eosinophilia, oligoclonal T cells, and elevated levels of IgE. When the CD4+ T cells in the mice were depleted, a reduction in their IgE levels resulted. Thus, Khiong et al concluded that these "memory mutant" mice may be a model for human Omenn syndrome, and that many manifestations of the murine disease were direct results of the RAG hypomorphism, whereas some were caused by malfunctions of their CD4+ T-cells.[4]

Artemis deficiency (with mutations in the Artemis protein that result in defective VDJ recombination) decreases both B and T cells and can be considered part of a subset of SCIDs. DNA ligase IV deficiency likewise results in defective circulating T- and B-cells and serum immunoglobulins.

Bloom syndrome, or congenital telangiectatic erythema, results from a mutation in the helicase enzyme called BLM RecQ. This mutation leads to defects in DNA repair and is characterized by an increased risk of malignancy and radiation sensitivity.

Defects in cytokine receptors and/or cytokine signaling (B cells are generally present but nonfunctional)

An extensive number of disorders with SCID manifestations belong to this category in which defects in cytokine receptors and/or cytokine signaling are present. Many cytokine receptors (eg, interleukin [IL], IL-2, IL-4, IL-7, IL-9, IL-15) share a common gamma chain, which is necessary for the normal signaling from the receptors after binding with their ligands.[5]

After binding of IL-2 to its receptor (ie, IL-2R), JAK3 is recruited to the cytoplasmic tail of the receptor and then phosphorylated. In turn, JAK3 phosphorylates a docking site for src homology-containing (SHC) signal transducer and activator of transcription (STAT) proteins. Subsequent phosphorylation and dimerization of STAT with its translocation into the nucleus results in gene transcription and/or activation.

The gene that encodes the gamma chain is located on band Xq13. Approximately 100 mutations have been described in this gene, resulting in an abnormal (two thirds of cases) or absent (one third of cases) gamma C-chain. The absence of the gamma-C chain or the presence of aberrant forms affect signaling events that are mediated via various cytokine receptors, thus explaining the multiple cell types that are affected in X-linked SCID, which include T, NK, and B cells.

X-linked SCID is characterized by the absence of T and NK cells but a normal number of dysfunctional B cells (T– B+ NK– SCID). The development of T cells is dependent on functional IL-7/IL-7R, and that of NK cells is dependent on functional IL-15/IL-15R, whereas the abnormalities of IL-2 and IL-4 pathways affect the function of B cells.

The gene encoding JAK3 is located on band 19p13. JAK3 deficiency results in a rare SCID syndrome that is also associated with absent T and NK cells but a normal number of dysfunctional B cells (T–B+NK–SCID).

The Wiskott-Aldrich syndrome protein (WASP) is encoded by a gene located on band Xp11.22–11.23. This protein has a dual role: (1) it affects immune cell motility and trafficking through its binding with CDC42H2 and rac, members of the Rho family of GTPases, which then results in changes in actin polymerization; and (2) it relays external signals into the nucleus. The mutated gene encodes a WASP that lacks the hydrophobic transmembrane domain and results in defective immune cell trafficking and motility. The abnormality affects all immune cells, including dendritic cells, macrophages, and B and T cells, leading to abnormal initiation and regulation of the immune response and, ultimately, to ineffective secondary lymphopoiesis.

In common variable immunodeficiency (CVID), mature B cells are normal in number and morphology, but they fail to differentiate into plasma cells because of defective interaction between the B and T cells, mostly caused by a T-cell defect. This defect is thought to be related to a decreased number and/or function of CD4+ T lymphocytes or, occasionally, to an increased number of CD8+ T lymphocytes; however, abnormal responses of B cells to many usual stimuli have also been identified in vitro.

In selective IgM deficiency, the underlying abnormality is a defect of helper T cells and excessive suppressor T-cell activity. The condition is characterized by a low IgM level. IgG) levels are normal, but the IgG response is usually decreased.

T-helper lymphocyte deficiency has been incriminated in the pathogenesis of transient hypogammaglobulinemia of infancy (THI) and immunodeficiency with thymoma.

Primary B-cell disorders result in a complete or partial absence of one or more immunoglobulin isotypes. Regardless of the primary cause, the clincal manifestations depend on the type and severity of the immunoglobulin deficiency and the association of cell-mediated immunodeficiency. In general, severe immunoglobulin deficiency results in recurrent infections with specific microorganisms at certain anatomic sites.

Immunoglobulins play a dual role in the immune response by recognizing foreign antigens and triggering a biologic response that culminates in the elimination of the antigen. Their role in the fight against bacterial infections has been recognized for many years. Emerging evidence from animal and clinical studies suggests a more important role for humoral immunity in the response to viral infections than was initially thought.

IgM plays a pivotal role in the primary immune response. It is the first immunoglobulin class produced in a primary response to an antigen. IgM binds the C1 component of complement and activates the classical pathway, leading to opsonization of antigens and cytolysis. Binding of IgM to the polyimmunoglobulin receptor brings IgM to mucosal surfaces.

IgG represents the major component of serum antibodies (ie, approximately 85%). By binding to the Fc receptors, they mediate many functions, including antibody-dependent cell-mediated cytotoxicity, phagocytosis, and clearance of immune complexes. IgG1 is the major component of the response to protein antigens (eg, antitetanus/diphtheria antibodies); IgG2 is produced in response to polysaccharide antigens (eg, antipneumococcal antibodies); and IgG3 seems to play an important role in the response to respiratory viruses.

Complement fixation and activation is carried out by IgG1, IgG3, IgM, and, to a lesser degree, IgG2. IgA and, to a lesser extent, IgM, produced locally and secreted by mucous membranes, are the major determinants of mucosal immunity.

IgG antibodies are the only immunoglobulin class that crosses the placenta. These placental antibodies provide the infant with effective humoral immunity during the first 7-9 months of life.

Deficiency of the expression of major histocompatibility complex (MHC) class I and II cellular proteins also commonly manifests in early infancy with classic expressions of SCID. Manifestations in affected patients indicate the crucial involvement of MHC proteins in the immune recognition of self and non-self.

In other B- and T-cell disorders, additional anomalies may predominate, and clinical manifestations suggestive of immunodeficiency may occur late in life. Patients with short-limbed skeletal dysplasia with cartilage-hair hypoplasia (CHH) can also have either a T-cell or combined defect.

Combined immunodeficiency due to caspase-8 deficiency presents as recurrent sinopulmonary bacterial infections, poor growth, lymphadenopathy and splenomegaly, asthma, and herpesvirus infection. Caspases are a family of proteases that play roles in signal transduction by inflammatory cytokine receptors (eg, IL-1 and IL-18) as well as in pathways leading to apoptosis. The percentage of CD4+ T cells is low (about 25% of lymphocytes) and the CD4/C8 ratio is 0.5. T cells showed decreased proliferation and IL-2 production in vitro with mitogens, and NK cell function is also impaired.

There are 2 autosomal recessive syndromes that indicate some interaction of the immune system with neurologic function: ataxia-telangiectasia (AT) and Nijmegen breakage syndrome (NBS). These involve various mutations of DNA proteins. AT is a rare, autosomal recessive, neurodegenerative disorder in which the diagnosis is based on the presence of both ataxia and telangiectasia; combined immunodeficiency can be quite variable in this condition. Other multisystemic manifestations of AT include motor impairments secondary to a neurodegenerative process, oculocutaneous telangiectasia, sinopulmonary infections, and hypersensitivity to ionizing radiation.

NBS is also an autosomal recessive chromosomal instability syndrome in which patients have increased susceptibility to infection or lymphatic tumor development due to defects in humoral and cellular immune functions. NBS is also characterized by microcephaly with growth retardation, normal or impaired intelligence, and birdlike facies. Nearly all patients with NBS are homozygous for the same founder mutation: deletion of 5 bp (657del5) in the NBS1 gene, which encodes the protein nibrin.

Both AT and NBS are associated with decreased circulating levels of T cells and often decreased levels of the IgA, IgE, and IgG subclasses, whereas circulating levels of B cells are normal.

Epidemiology

Frequency

United States

The accurate incidence of SCID in the United States is unknown, but it has been estimated to be in 1 per 50,000-100,000 births across all ethnic groups. A postulated reason for the lack of exact epidemiologic information is that infants with SCID may die of infections without having been diagnosed with the condition.

With implementation of SCID newborn screening in unbiased populations, Kwan et al reported that 1 in 58,000 infants (95% CI 1/46,000–80,000) are born with SCID or leaky SCID (ie, forms of SCID, such as Omenn syndrome, characterized by normal or elevated levels of nonfunctional T cells, in contrast to the low or absent T cell counts of typical SCID). That prevalence rate is nearly twice the previous estimates based on population data or experience of centers performing hematopoietic cell transplantation therapy for SCID.[6]

The approximate frequency of the most common forms of SCID is as follows:

The incidence of reticular dysgenesis and CHH are less than 1% each. In approximately 14% of cases, the etiology remains unknown.[7]

International

Estimates for Europe are thought to approximate those in the United States. CHH may be more frequent in Finland. SCID is underreported, but several countries now maintain registries of patients with primary immunodeficiency diseases.

The estimated prevalence of SCID in specific countries is as follows[8] :

Mortality/Morbidity

SCID is a devastating disease with a high risk of early death in infancy or childhood: a large number of patients die during their first year of life, and most do not survive beyond their second year.

The condition is notable for recurrent failure to thrive and common infections (eg otitis media, diarrhea, mucocutaneous candidiasis). Moreover, if infants are not diagnosed by age 6 months, opportunistic infections follow, especially Pneumocystis jirovecii pneumonia and invasive fungal infections, and mortality may ensue from infections with common viruses, such as the following[7, 9] :

Race-, sex-, and age-related demographics

Although there is no racial predilection for combined B-cell and T-cell disorders, some forms of combined immunodeficiency have been reported more in some ethnic groups, such as the following[7] :

The disorders associated with the X chromosome typically manifest only in males, whereas females are carriers. Approximately 50% of SCID cases are X-linked.

Most patients with these disorders become symptomatic with recurrent infections, failure to thrive, or both in the first months of life.

History

The clinical hallmarks of severe combined immunodeficiency (SCID) are repetitive and frequent bacterial, viral, and fungal infections that persist despite standard medical treatment. These result from the profound degree of immune compromise in SCID.

Patients with primary T-cell deficiency SCID begin having infections soon after birth (ie, age 3-4 mo) compared with those that have pure B-cell disorders, who do not have an increased incidence of bacterial infections until 7-9 months after birth, when placental antibodies fall to undetectable levels.

Clinicians should focus attention on the family history, site of infection, type of microorganisms, and any adverse reactions to transfusion of blood products, which may provide clues to the significance and type of immune deficiency. It is also important to inquire about consanguineous relationships because consanguinity increases the risk of immune disorders that have autosomal recessive inheritance patterns (eg, some forms of SCID or chronic granulomatous disease [CGD]). In addition, a careful family history of risk factors for human immunodeficiency virus (HIV) should be obtained to rule out secondary forms of immunodeficiency.

Upper and lower respiratory tract infections, skin infections, meningitis, bacteremias, and abscesses are common in persons with B-cell disorders. Pneumonia with Pneumocystis jirovecii or cytomegalovirus (CMV), disseminated bacillus Calmette-Guerin (BCG) infection,[11] or atypical mycobacterial infection and recurrent or persistent skin candidiasis are suggestive of T-cell disorders or SCID. Diarrhea with failure to thrive in children with SCID is usually related to infections with viruses such as rotaviruses and adenoviruses. Although antibody deficiency is associated with recurrent encapsulated bacteria infections, T-cell disorders or SCID are associated with opportunistic infections with fungi, viruses, or intracellular bacteria.

Reactions to blood products or vaccines should raise the suggestion of an underlying immunodeficiency, particularly IgA deficiency. Transfusion with blood products can result in significant graft versus host disease (GVHD) in SCID patients.

After a detailed inquiry, a SCID disorder should be suspected if the patient falls into one of the following groups:[12, 13, 14, 15]

X-linked severe combined immunodeficiency

X-linked severe combined immunodeficiency (XSCID) is by far the most common form of SCID, accounting for almost 50% of cases.[16]  As the affected gene is located in the X chromosome (X13q band), the disease is limited to males. Because of a defective common gamma chain (a component of cytokine receptors for interleukin-2 [IL-2], IL-4, IL-7, IL-9, and IL-15), signal transduction cannot proceed normally, which results in SCID characterized by absent T and NK cells and dysfunctional B cells. The phenotype is T–B+NK–.

Infections begin in the first months of life, affecting the upper and lower respiratory tracts, gastrointestinal tract, and skin, whereas X-linked agammaglobulinemia (XLA) does not manifest clinically until the second half of an infant's first year of life. Persistent opportunistic infections with Candida albicans or P jirovecii and viral infections with varicella-zoster virus, CMV, and Epstein-Barr virus are common. The risk of GVHD is high in these patients because of their inability to reject foreign antigens

X-linked immunodeficiency with hyper IgM syndrome 

X-linked immunodeficiency with hyper IgM syndrome (XHM) is a part of the hyper-IgM syndromes that includes a group of disorders characterized by recurrent bacterial infections and low serologic levels of IgG, IgA, and IgE, with relatively elevated levels of IgM. XHM affects only boys and is the result of mutations in the gene that encodes the CD40 ligand (CD40L or CD154) located on chromosome X. Clinical manifestations of XHM are as follows:

Adenosine deaminase (ADA) deficiency

ADA is an enzyme of the purine salvage pathway. Deficiency leads to the accumulation of dATP and 2'-deoxyadenosine. dATP is lymphocytotoxic because of its ability to inhibit DNA synthesis via inhibition of ribonucleotide reductase. The nucleoside 2'-deoxyadenosine inhibits the enzyme S-adenosyl-L-homocysteine (SAH) hydrolase, which results in accumulation of SAH, a potent inhibitor of all cellular methylation reactions. Both B and T cells are affected. The phenotype is T–B–NK–.

ADA deficiency is an autosomal recessive disorder in which the age at presentation varies. Failure of the immune system is progressive and may not fully manifest in certain individuals until adulthood.

This disease has the same symptoms of XSCID, that is, recurrent infections, persistent opportunistic infections, and GVHD susceptibility. Clinically, ADA deficiency differs from XSCID by (1) the presence of skeletal and chest wall abnormalities involving the vertebral bodies and the chondrocostal junctions and (2) the possible presence of thymic differentiation with rare Hassall concentric corpuscles.

JAK3 deficiency

JAK3 is an intracellular enzyme that is activated as a result of the binding of cytokines with their cognate receptors. The gene encoding JAK3 is located on band 19p13, and the disorder is autosomal recessive. The phenotype is T–B+NK–.

The symptoms of this condition are similar to those observed in persons with XSCID and include upper and lower respiratory tract infections, persistent infections with opportunistic microorganisms, and increased susceptibility to GVHD.

RAG1 and RAG2 deficiency

In patients deficient in the RAG proteins 1 and 2, the lymphocytes cannot rearrange the antigen receptors, thus leading to B- and T-lymphocyte deficiency. Phenotypically, the numbers of B and T cells are decreased, whereas the number of NK cells is normal.

Clinically, these patients present with increased susceptibility to infection with encapsulated and intracellular bacteria, viruses, and fungi. On laboratory studies, this syndrome is characterized by high serum IgE levels, decreased levels of the other immunoglobulins, and hypereosinophilia.

RAG1 deficiency is observed in patients with cartilage-hair hypoplasia (CHH) and is characterized by the following:

CHH

CHH manifests in early infancy as chronic diarrhea, failure to thrive, and an erythematous rash with desquamation. Hepatosplenomegaly is common. Patients die in the first few months of life unless successful allogeneic bone marrow transplantation is performed.

Reticular dysgenesis

Reticular dysgenesis is a rare disorder that is characterized by an almost complete lack of granulocytes and lymphocytes. Most patients die in early infancy or the newborn period because of severe and overwhelming infection. The molecular basis of the disease is not known

Wiscott-Aldrich syndrome

Wiskott-Aldrich Syndrome (WAS) is an X-linked recessive disorder resulting from mutation in a gene that encodes for the WAS protein (WASP). WASP is a key regulator of actin polymerization in hematopoietic cells. Structural studies of the WASP protein have identified 5 domains that are involved in signaling, cell locomotion, and immune synapse formation. WASP regulates nuclear factor kappaB (NF-KB) activity by promoting the nuclear translocation of NF-KB. In addition, WASP plays not only an important role in lymphoid development, but also in the maturation of myeloid monocytic cells.

Clinically, the syndrome is characterized by the triad of thrombocytopenic purpura, eczema, and increased susceptibility to infections. Symptoms begin in the first year of life, with recurrent upper and lower respiratory tract infections with encapsulated bacteria. P jirovecii pneumonia and herpes infections become a problem later in life

Most patients die of serious infections at approximately age 11 years. If these patients survive to adulthood, they are at high risk for autoimmune diseases, such as cytopenias and vasculitis, and for cancer, particularly non-Hodgkin lymphomas.

The discovery of the Wiskott-Aldrich gene made possible the identification of carriers of the gene in families of WAS patients with an incomplete syndrome. Some of these patients have only the thrombocytopenia (X-linked thrombocytopenia) with no skin involvement or immunodeficiency despite inheriting the same gene mutation.

Physical

The physical examination may identify nonspecific signs of acute or chronic infections and those more specifically related to certain disease entities. Consider the following:

Causes

SCID disorders are the result of specific genetic alterations in key regulators of B-cell, T-cell, and/or natural killer (NK)-cell activation, proliferation, or differentiation. The genetic alterations have been identified in the following disorders, which has led to the investigation of gene therapy as an attractive intervention to treat such conditions:

Approach Considerations

Screening

In the United States as of December 2018, all 50 states, as well as the District of Columbia, the Navajo Nation, and Puerto Rico, conduct population-wide newborn screening (NBS) for SCID.[19]  T-cell receptor excision circles (TRECs), a biomarker for T lymphopoiesis, can be measured by polymerase chain reaction (PCR) using DNA isolated from infant dried blood spots (DBS).[6, 10]  

A systematic review of the diagnostic performance of published algorithms for TREC-based NBS for SCID concluded that a using a TREC cutoff value of maximal 25 TRECs/μl and incorporating the collection of a repeat DBS from neonatal intensive care unit patients with an abnormal screening result in the screening algorithm would be most effective in screening newborns for primary immunodeficiencies with T cell lymphopenia.[20]

Laboratory Studies

The diagnosis of SCID should be suspected in children with any of the following conditions:

Patients with suspected SCID require complete evaluation of specific humoral and cellular immunity, which includes measurement of immunoglobulin levels, antibody titers, lymphocyte subsets, and assessment of T-cell function. This can be done via evaluating the responses to mitogens in vitro.

The probable diagnosis of SCID is based on the following:

Levels of serum immunoglobulin are determined by serum protein electrophoresis.

Quantitative methods are used for the precise measurement of each immunoglobulin isotype. Enzyme-linked immunosorbent assays (ELISAs) are used for IgE quantitation.

Compare values to age-standardized reference ranges for each laboratory. The following are examples of values that are used for the adult population:

Antibody response after immunization may be absent.

The absence of isohemagglutinins is a significant finding that is suggestive of an immunoglobulin production problem. Evaluate IgM antibodies to A and B blood group antigens (isohemagglutinins) if the other test findings are within reference ranges and the patient is unable to mount a response to specific antigens.

Peripheral blood lymphocyte levels should be measured.

Lymphocyte phenotyping using flow cytometry analysis is the next step. The absolute number of B-cells, T-cells, and natural killer (NK) cells is more useful than percentages.

Measuring T-lymphocyte numbers and function may be necessary. Lymphocyte activation (CD45 RA/RO isoformic antigens) and T-cell receptor phenotype (TCR ab/gd lineage) determination may provide additional information regarding the type of immunodeficiency. For example, Omenn syndrome is characterized by a high number of T cells carrying TCRgd or CD45+. Determination of the helper (CD4) to suppressor (CD8) T-cell ratio is sometimes useful.

Cutaneous delayed-type hypersensitivity testing is used to evaluate the anamnestic response of cellular immunity to previously encountered antigens.

Results in specific disorders

XHM:

ADA deficiency:

RAG1 and RAG2 deficiency:

Imaging Studies

Chest radiography

Sometimes, recurrent or chronic infections may lead to abnormal chest radiographic findings, such as interstitial infiltrates, bronchiectasis, emphysema, and scarring. Note: Normal chest radiographic findings do not exclude the presence of structural abnormalities.

Absence of a thymic shadow is a very common finding in SCID. Patients with DiGeorge syndrome and other T-cell defects may also lack thymic tissue. However, the presence of thymic tissue does not exclude SCID. For example, patients with SCID who have mutations in ZAP70 or CD3 typically have normal-sized thymuses.

Chest radiographs in patients with ADA deficiency typically show inferior scapular angle squaring and spurring and costochondral cupping. Verhagen et al reported that these findings can reliably differentiate ADA deficiency from other forms of SCID, in children younger than approximately 7 months of age.[21]

Other Tests

For a prenatal diagnosis, restriction fragment length polymorphism (RFLP)can help detect genetic defect carriers of XHM, WAS, and ADA deficiency using fetal blood, amnion cells, or chorionic villus tissue. Umbilical cord blood can be used in the prenatal diagnosis of some of these disorders.

T cells are absent in persons with XSCID. B cells and T cells are absent in patients with autosomal recessive SCID. "Bald" lymphocytes found on scanning electron microscopy are diagnostic of WAS. Red blood cell ADA is decreased in fetuses with ADA deficiency.

ADA deficiency can be evaluated by demonstrating the following:

In AT, chromosomal karyotyping should reveal reciprocal translocations between chromosomes 7 and 14. Chromosomal instability testing is done to confirm AT and NBS to assess spontaneous and induced breakage. Diagnostic findings are absence or dysfunction of the ATM protein and mutations in the ATM gene.

Procedures

See the list below:

Approach Considerations

Intravenous immunoglobulin (IVIG) replacement therapy may benefit patients with combined immunodeficiencies, such as severe combined immunodeficiency (SCID), X-linked immunodeficiency with hyper IgM syndrome (XHM), Good syndrome, and Wiskott-Aldrich syndrome (WAS). Appropriate supportive care, such as early identification of opportunistic infections or nutritional support, are necessary.

In WAS, other than prophylactic antibiotics and IVIG, splenectomy for thrombocytopenia and platelet transfusion in acute life-threatening bleeding can be used.

Note: Do not immunize these patients with live attenuated vaccines. Focus efforts on the treatment of infections, allergic reactions, and autoimmune and gastrointestinal diseases. Aggressive and prolonged antibiotic therapy covering Streptococcus pneumoniae and Haemophilus influenzae is indicated. Prophylactic antibiotic therapy has been recommended for patients with frequent infections. A course of metronidazole may result in dramatic improvement of the diarrhea and, to a certain extent, of malabsorption syndrome in these patients. Prophylactic antibiotic therapy may significantly decrease the incidence of infections.

Patients with adenosine deaminase (ADA) deficiency may benefit from adenosine deaminase enzyme replacement therapy with pegademase or elapegademase. The maximum effect on immunologic function does not occur for several months.

Allogeneic bone marrow or hematopoietic stem cell transplantation (HSCT) may be helpful for patients with SCID. Survival rates in these previously fatal conditions are around 90% in some case series.

This treatment strategy is highly successful when a human leukocyte antigen (HLA)–matched sibling donor is available; if such a donor is not available, however, few therapeutic options exist. Gene-modified, autologous bone marrow transplantation can circumvent the severe immunologic complications that occur when a related HLA-mismatched donor is used and thus represents an attractive alternative (see below). 

Allogeneic bone marrow transplantation has become the standard of care for certain patients with SCIDs (eg, X-linked severe combined immunodeficiency [XSCID], ADA deficiency). Patients with other immunodeficiency syndromes may benefit from bone marrow transplantation or HSCT, including those with WAS or XHM.

Many groups are exploring the potential benefits of HSCT based on alternative donors. Umbilical cord blood stem cell transplantation (UCBSCT) offers several advantages, including ready availability of the unit, a lower risk of transmitting viral diseases, no risk to the donor, and a lower risk of GVHD even in the absence of a perfect HLA match.

Another possibility for patients without a suitable sibling donor is a matched unrelated donor (MUD) HSCT. But in clinical practice, this therapy is limited due to high rates of graft versus host disease (GVHD) and transplant-related mortality.

Identification of a suitable MUD has been facilitated by recent advances, including the following: 

  1.  Continuous growth of volunteer donors worldwide
  2. High-resolution molecular techniques for HLA typing, which permits a better selection of donors
  3. Advances in critical care that have resulted in a significant decrease in MUD-HSCT transplant–related mortality and an increase in the survival of SCID infants who are severely infected at the time of diagnosis.

Early diagnosis before the development of permanent lung and liver damage and referral to a specialized center for bone marrow transplantation/HSCT are essential for therapeutic success.

Bertrand et al reported on a European experience with 178 patients in 18 centers who were treated with HLA, nonidentical, T-cell–depleted bone marrow transplantation.[23]  With a median follow-up of 57 months, disease-free survival was shown to be significantly better for patients with B-positive SCID (60%) than for patients with B-negative SCID (35%).[23]

Buckley et al found that the survival rate was not affected by the genetic type, but it was affected by race (ie, more white patients than black or Hispanic patients survived [P < 0.001]) and sex (all girls survived [P = 0.047]).[24]

Another report noted the inefficacy of bone marrow transplantation in correcting Job syndrome.[25]

Gene therapy

Despite the success that has been seen in some SCID patients treated with bone marrow transplantation, other patients experince failure to restore B-cell function or failure or rejection of the graft over time. A novel alternative strategy to circumvent graft failure/rejection is the use of gene transfer into autologous stem cells using retroviruses.

Gene therapy is a viable therapeutic option; advances in biotechnology have enabled the performance of this highly complex treatment for several immunodeficiency syndromes.[26, 27]

Cavazzana-Calvo et al published reports of the successful results of gene therapy for SCID-X1 disease in 2 children, opening new horizons for the future of these patients.[5]  This therapy resulted in complete immune reconstitution of the lymphoid system, with T-, B-, and NK-cell counts comparable to age-matched controls.[5]  An update on these 2 patients by the same authors and a report on 3 others confirmed the previous results.

Patients with ADA deficiency were the first to be enrolled in gene therapy trials. Since 2000, over 100  patients with ADA-SCID have been treated with gamma-retrovirus or lentivirus-mediated autologous hematopoietic stem cell gene therapy (HSC-GT), and the excellent safety and efficacy observed in these cases has established HSC-GT as an equal alternative to HSCT for these patients.[28]

In addition to ADA deficiency, , SCID-X1, WAS, RAG1 deficiency, and CD3 deficiency have been successfully treated with gene-modified autologous HSC-GT. In this technique, RNA viruses are the vectors most commonly used to introduce genetic information into hematopoietic stem cells and/or progenitor cells. It has been demonstrated that several T-cell precursors that carry a wild-type sequence of the disease-causing gene or a less harmful mutation can mature into functional mature T cells that provide adequate immunity. 

Consultations

Consultations should be obtained with specialists from the following specialties:

Diet

In view of the presence of chronic diarrhea, patients often require enteral or parenteral supplementation.

Activity

Physical activity should be encouraged. Patients may need isolation to decrease the risk of common viral and bacterial infections, such as avoiding crowded places. Strict hygienic practices are important.

Medication Summary

The goals of pharmacotherapy are to reduce morbidity and to prevent complications.

Immune globulin, intravenous (Gamimune, Gammagard, Sandoglobulin, Gammar-P)

Clinical Context:  Provide an immediate rise of antibodies that have a proven protective effect against bacterial and viral infection (passive immunity). Because antibodies are not produced by the host, these products must be readministered monthly. This treatment may increase CSF IgG (10%).

Class Summary

Blood products/immunoglobulins provide immediate passive immunity. These agents can be used as replacement therapy in patients with antibody-deficiency states.

Elapegademase (Revcovi, Elapegademase-lvlr)

Clinical Context:  Elapegademase a recombinant adenosine deaminase based on bovine amino acid sequence and conjugated to PEG. It is indicated for treatment of ADA severe combined immune deficiency (ADA-SCID) in pediatric and adult patients.

Pegademase bovine (Adagen)

Clinical Context:  ADA is an enzyme of the purine salvage pathway that is responsible for adenosine and deoxyadenosine deamination to inosine and deoxyinosine, respectively. ADA deficiency leads to accumulation of the metabolites dATP and 2'-deoxyadenosine, both of which are toxic to lymphocytes.

Treatment is indicated in patients with SCID secondary to ADA deficiency whose conditions proved refractory to bone marrow transplantation or who are not candidates for transplantation. Individualize therapy (based on plasma levels) to achieve the following: trough plasma levels of 15-35 mmol/h/mL and a decline in erythrocyte dATP to < 0.005-0.015 mmol/mL packed erythrocytes or to < 1% of total erythrocyte adenine nucleotide content (ATP + dATP). Plasma levels >35 mmol/h/mL are not associated with additional clinical benefit. This treatment has no role in preparatory regimen for bone marrow transplantation.

Class Summary

Adenosine deaminase (ADA) enzyme replacement can reduce potentially serious, life-threatening infections in ADA deficient patients.

Further Outpatient Care

Regularly monitor the following in patients with severe combined immunodeficiency:

Imunoglobulin trough levels of 400 mg/dL or higher are considered satisfactory. Occasionally, levels greater than 500 mg/dL are required to clear certain viral infections, such as enterovirus meningoencephalitis. Trough levels may need to be titrated in individual patients to determine the level that is needed to prevent recurrent infection.

If abnormalities are identified on liver function tests, imaging studies of the liver and biliary tree are necessary to exclude malignancies or sclerosing cholangitis (the latter is observed in patients with X-linked immunodeficiency with hyper IgM syndrome [XHM]).

Inpatient & Outpatient Medications

See the list below:

Transfer

See the list below:

Deterrence/Prevention

See the list below:

Complications

See the list below:

Prognosis

See the list below:

Patient Education

See the list below:

What is severe combined immunodeficiency (SCID)?How has the understanding of severe combined immunodeficiency (SCID) evolved?What is the pathophysiology of severe combined immunodeficiency (SCID)?What are the pathophysiologic pathways of severe combined immunodeficiency (SCID)?What is the role of purine metabolites in the pathophysiology of severe combined immunodeficiency (SCID)?What is the role of RAG1 and RAG2 proteins in the pathophysiology of severe combined immunodeficiency (SCID)?What is the role of cytokine receptors in the pathophysiology of severe combined immunodeficiency (SCID)?What is the US prevalence of severe combined immunodeficiency (SCID)?What is the global prevalence of severe combined immunodeficiency (SCID)?What is the mortality and morbidity associated with severe combined immunodeficiency (SCID)?What are the racial predilections of severe combined immunodeficiency (SCID)?What are the sexual predilections of severe combined immunodeficiency (SCID)?At what age does severe combined immunodeficiency (SCID) first present?Which clinical history findings are characteristic of X-linked immunodeficiency with hyper IgM syndrome (XHM)?Which clinical history findings are characteristic of Wiskott-Aldrich syndrome (WAS)?Which clinical history findings are characteristic of severe combined immunodeficiency (SCID)?Which clinical history findings are characteristic of X-linked severe combined immunodeficiency (XSCID)?Which clinical history findings are characteristic of adenosine deaminase (ADA) deficiency?Which clinical history findings are characteristic of JAK3 deficiency?Which clinical history findings are characteristic of RAG1 and RAG2 deficiency?Which clinical history findings are characteristic of cartilage-hair hypoplasia (CHH)?Which clinical history findings are characteristic of reticular dysgenesis?Which physical findings are characteristic of severe combined immunodeficiency (SCID)?What causes severe combined immunodeficiency (SCID)?How is severe combined immunodeficiency (SCID) differentiated from pure B-cell disorders?What are the differential diagnoses for Severe Combined Immunodeficiency (SCID)?How is severe combined immunodeficiency (SCID) screened in newborns?Which lab findings are characteristic of ADA deficiency?Which lab findings are characteristic of RAG1 and RAG2 deficiency?How is severe combined immunodeficiency (SCID) diagnosed?Which lab findings are characteristic of X-linked immunodeficiency with hyper IgM syndrome (XHM)?What is the role of chest radiography in the workup of severe combined immunodeficiency (SCID)?How is severe combined immunodeficiency (SCID) diagnosed prenatally?How is ADA deficiency diagnosed?How is ataxia-telangiectasia (AT) diagnosed?Which procedures are performed in the workup of severe combined immunodeficiency (SCID)?How is severe combined immunodeficiency (SCID) treated?What is the role of gene therapy in the treatment of severe combined immunodeficiency (SCID)?Which specialist consultations are beneficial to patients with severe combined immunodeficiency (SCID)?Which dietary modifications are used in the treatment of severe combined immunodeficiency (SCID)?Which activity modifications are used in the treatment of severe combined immunodeficiency (SCID)?What is the role of medications in the treatment of severe combined immunodeficiency (SCID)?Which medications in the drug class Metabolic Enzymes are used in the treatment of Severe Combined Immunodeficiency (SCID)?Which medications in the drug class Blood Products/Immunoglobulins are used in the treatment of Severe Combined Immunodeficiency (SCID)?What is included in the long-term monitoring of severe combined immunodeficiency (SCID)?How is IVIG therapy administered for the treatment of severe combined immunodeficiency (SCID)?When is patient transfer indicated for the treatment of severe combined immunodeficiency (SCID)?How are infections prevented in severe combined immunodeficiency (SCID)?What are the possible complications of IVIG therapy for severe combined immunodeficiency (SCID)?What are the possible complications of severe combined immunodeficiency (SCID)?What is the prognosis of severe combined immunodeficiency (SCID)?What is included in patient education about severe combined immunodeficiency (SCID)?

Author

Francisco J Hernandez-Ilizaliturri, MD, Professor of Medicine, Department of Medical Oncology, Associate Professor of Immunology, Department of Immunology, Chief, Lymphoma and Myeloma Section, Director, The Lymphoma Translational Research Program, Roswell Park Cancer Institute, University of Buffalo State University of New York School of Medicine and Biomedical Sciences

Disclosure: Nothing to disclose.

Coauthor(s)

David Claxton, MD, Professor of Medicine, Department of Internal Medicine, Section of Hematology-Oncology, Hershey Medical Center, Pennsylvania State University College of Medicine

Disclosure: Nothing to disclose.

Issam Makhoul, MD, Associate Professor, Department of Medicine, Division of Hematology/Oncology, University of Arkansas for Medical Sciences

Disclosure: Nothing to disclose.

Mohammad Muhsin Chisti, MD, FACP, Assistant Professor of Hematology and Oncology, Medical Director of Research, Karmanos Cancer Institute, Wayne State University School of Medicine

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Marcel E Conrad, MD, Distinguished Professor of Medicine (Retired), University of South Alabama College of Medicine

Disclosure: Partner received none from No financial interests for none.

Chief Editor

Emmanuel C Besa, MD, Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Disclosure: Nothing to disclose.

Acknowledgements

James O Ballard, MD Kienle Chair for Humane Medicine, Professor, Departments of Humanities, Medicine, and Pathology, Division of Hematology/Oncology, Milton S Hershey Medical Center, Pennsylvania State University College of Medicine

James O Ballard, MD is a member of the following medical societies: Alpha Omega Alpha, American College of Physicians, and American Society of Hematology

Disclosure: Nothing to disclose.

References

  1. Justiz Vaillant AA, Mohseni M. Severe Combined Immunodeficiency. 2020 Jan. [View Abstract]
  2. Stray-Pedersen A, Sorte HS, Samarakoon P, Gambin T, Chinn IK, et. al. Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders. J Allergy Clin Immunol. 2017 Jan;139(1):232-245. [View Abstract]
  3. Cavazzana-Calvo M, Fischer A. Gene therapy for severe combined immunodeficiency: are we there yet?. J Clin Invest. 2007 Jun. 117(6):1456-65. [View Abstract]
  4. Khiong K, Murakami M, Kitabayashi C, et al. Homeostatically proliferating CD4 T cells are involved in the pathogenesis of an Omenn syndrome murine model. J Clin Invest. 2007 May. 117(5):1270-81. [View Abstract]
  5. Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science. 2000 Apr 28. 288(5466):669-72. [View Abstract]
  6. Kwan A, Abraham RS, Currier R, et al. Newborn screening for severe combined immunodeficiency in 11 screening programs in the United States. JAMA. 2014 Aug 20. 312 (7):729-38. [View Abstract]
  7. Sinha S, Schwartz RA. Severe combined immunodeficiency. Medscape Reference. Updated August 21, 2006.
  8. Bonilla FA, Geha RS. 2. Update on primary immunodeficiency diseases. J Allergy Clin Immunol. 2006 Feb. 117(2 suppl mini-primer):S435-41. [View Abstract]
  9. Cachafeiro T, Escobar G, Bakos L, Bakos R. Chronic cutaneous cytomegalovirus infection in a patient with severe combined immunodeficiency syndrome. Br J Dermatol. 2013 Sep 6. [View Abstract]
  10. Kwan A, Puck JM. History and current status of newborn screening for severe combined immunodeficiency. Semin Perinatol. 2015 Apr. 39 (3):194-205. [View Abstract]
  11. Bacalhau S, Freitas C, Valente R, Barata D, Neves C, Schäfer K, et al. Successful Handling of Disseminated BCG Disease in a Child with Severe Combined Immunodeficiency. Case Report Med. 2011. 2011:527569. [View Abstract]
  12. Verbsky JW, Baker MW, Grossman WJ, Hintermeyer M, Dasu T, Bonacci B, et al. Newborn Screening for Severe Combined Immunodeficiency; The Wisconsin Experience (2008-2011). J Clin Immunol. 2011 Nov 10. [View Abstract]
  13. Somech R, Lev A, Simon AJ, Korn D, Garty BZ, Amariglio N, et al. Newborn screening for severe T and B cell immunodeficiency in Israel: a pilot study. Isr Med Assoc J. 2013 Aug. 15(8):404-9. [View Abstract]
  14. Kelly BT, Tam JS, Verbsky JW, Routes JM. Screening for severe combined immunodeficiency in neonates. Clin Epidemiol. 2013 Sep 16. 5:363-369. [View Abstract]
  15. Rozmus J, Junker A, Thibodeau ML, Grenier D, Turvey SE, Yacoub W, et al. Severe Combined Immunodeficiency (SCID) in Canadian Children: A National Surveillance Study. J Clin Immunol. 2013 Oct 12. [View Abstract]
  16. Zhang C, Zhang ZY, Wu JF, Tang XM, Yang XQ, Jiang LP, et al. Clinical characteristics and mutation analysis of X-linked severe combined immunodeficiency in China. World J Pediatr. 2011 Nov 21. [View Abstract]
  17. Levy J, Espanol-Boren T, Thomas C, et al. Clinical spectrum of X-linked hyper-IgM syndrome. J Pediatr. 1997 Jul. 131(1 pt 1):47-54. [View Abstract]
  18. Ridanpaa M, van Eenennaam H, Pelin K, et al. Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell. 2001 Jan 26. 104(2):195-203. [View Abstract]
  19. IDF SCID Newborn Screening Campaign. Immune Deficiency Foundation. Available at https://primaryimmune.org/idf-advocacy-center/idf-scid-newborn-screening-campaign. Accessed: May 28, 2020.
  20. van der Spek J, Groenwold RH, van der Burg M, van Montfrans JM. TREC Based Newborn Screening for Severe Combined Immunodeficiency Disease: A Systematic Review. J Clin Immunol. 2015 May. 35 (4):416-30. [View Abstract]
  21. Verhagen MV, Trevisan V, Adu J, Owens CM, Booth C, Calder A. Chest Radiographs for Distinguishing ADA-SCID from Other Forms of SCID. J Clin Immunol. 2020 Feb. 40 (2):259-266. [View Abstract]
  22. Chin T, Alonazi N. B-cell and T-cell combined disorders. Medscape Reference. Updated April 5, 2007.
  23. Bertrand Y, Landais P, Friedrich W, et al. Influence of severe combined immunodeficiency phenotype on the outcome of HLA non-identical, T-cell-depleted bone marrow transplantation: a retrospective European survey from the European Group for Bone Marrow Transplantation and the European Society for Immunodeficiency. J Pediatr. 1999 Jun. 134(6):740-8. [View Abstract]
  24. Buckley RH, Schiff SE, Schiff RI, et al. Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency. N Engl J Med. 1999 Feb 18. 340(7):508-16. [View Abstract]
  25. Gennery AR, Flood TJ, Abinun M, Cant AJ. Bone marrow transplantation does not correct the hyper IgE syndrome. Bone Marrow Transplant. 2000 Jun. 25(12):1303-5. [View Abstract]
  26. Castiello MC, Scaramuzza S, Pala F, Ferrua F, Uva P, Brigida I, et al. B-cell reconstitution after lentiviral vector-mediated gene therapy in patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol. 2015 Sep. 136 (3):692-702.e2. [View Abstract]
  27. Fischer A, Hacein-Bey-Abina S. Gene therapy for severe combined immunodeficiencies and beyond. J Exp Med. 2020 Jan 6. 217 (2):[View Abstract]
  28. Kohn DB, Hershfield MS, Puck JM, Aiuti A, Blincoe A, Gaspar HB, et al. Consensus approach for the management of severe combined immune deficiency caused by adenosine deaminase deficiency. J Allergy Clin Immunol. 2018 Sep 5. [View Abstract]
  29. Atluri S, Neville K, Davis M, et al. Epstein-Barr-associated leiomyomatosis and T-cell chimerism after haploidentical bone marrow transplantation for severe combined immunodeficiency disease. J Pediatr Hematol Oncol. 2007 Mar. 29(3):166-72. [View Abstract]
  30. Husain M, Grunebaum E, Naqvi A, et al. Burkitt's lymphoma in a patient with adenosine deaminase deficiency-severe combined immunodeficiency treated with polyethylene glycol-adenosine deaminase. J Pediatr. 2007 Jul. 151(1):93-5. [View Abstract]
  31. Hadzic N, Pagliuca A, Rela M, et al. Correction of the hyper-IgM syndrome after liver and bone marrow transplantation. N Engl J Med. 2000 Feb 3. 342(5):320-4. [View Abstract]
  32. Zhu Q, Watanabe C, Liu T, et al. Wiskott-Aldrich syndrome/X-linked thrombocytopenia: WASP gene mutations, protein expression, and phenotype. Blood. 1997 Oct 1. 90(7):2680-9. [View Abstract]
  33. Picard C, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, Conley ME, et al. Primary Immunodeficiency Diseases: an Update on the Classification from the International Union of Immunological Societies Expert Committee for Primary Immunodeficiency 2015. J Clin Immunol. . 2015 Nov;35(8):696-726. [View Abstract]
  34. Shearer WT, Dunn E, Notarangelo LD, Dvorak CC, Puck JM, Logan BR, et al. Establishing diagnostic criteria for severe combined immunodeficiency disease (SCID), leaky SCID, and Omenn syndrome: the Primary Immune Deficiency Treatment Consortium experience. J Allergy Clin Immunol. 2014 Apr;133(4):1092-8. [View Abstract]
  35. Wiekmeijer AS, Pike-Overzet K, IJspeert H, Brugman MH, Wolvers-Tettero IL, Lankester AC, et al. Identification of checkpoints in human T-cell development using severe combined immunodeficiency stem cells. J Allergy Clin Immunol. 2016 Feb;137(2):517-526. [View Abstract]
  36. Tagliaferri L, Kunz JB, Happich M, Esposito S, Bruckner T, Hübschmann D, et al. Newborn screening for severe combined immunodeficiency using a novel and simplified method to measure T-cell excision circles (TREC). Clin Immunol. 2017 Feb;175:51-55. [View Abstract]
  37. Rezaei N, Aghamohammadi A, Notarangelo L D. Primary Immunodeficiency Diseases. Springer Berlin Heidelberg; December 2016. 83-182.
Pathophysiology Cells Affected Inheritance Genes Involved
Premature cell deathT, B, NKAR ADA
Defective cytokine–dependent survival signalingT, NKAR



γ c type-XL



JAK3, IL7RA (T cells only), γ c
Defective V(D)J rearrangementT, BARRAG1, RAG2, Artemis
Defective pre-TCR and TCR signalingTAR CD3 δ, CD3 ζ, CD3 ε,



CD45



AR = autosomal recessive; JAK3 =Janus tyrosine kinase 3; RAG1, RAG2 = recombinase activating gene 1 and 2, respectively; TCR = T-cell receptor; XL = X-linked; V(D)J = variable diversity joining.