Bookshelf » GeneReviews » X-Linked Severe Combined Immunodeficiency
 
gene
GeneReviews
PagonRoberta A
BirdThomas C
DolanCynthia R
SmithRichard JH
StephensKaren
University of Washington, Seattle2009
geneticspublic health

GeneTests Home Page About GeneTests Search GeneReviews on the GeneTests web site Laboratory Directory Clinic Directory Educational Materials Illustrated Glossary

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

X-Linked Severe Combined Immunodeficiency
[SCID, X-Linked; SCIDX1; X-SCID]

Joie Davis, APRN, BC, APNG
Genetics and Molecular Biology Branch
National Human Genome Research Institute
National Institutes of Health
Pediatric Nurse Practitioner and Study Coordinator with the X-SCID population
Jennifer M Puck, MD
Chief, Genetics and Molecular Biology Branch
National Human Genome Research Institute
National Institutes of Health
Head, Immunologic Genetics Section
12122005x-scid
Initial Posting: August 26, 2003.
Last Update: December 12, 2005.

*

*

*

Summary

Disease characteristics. X-linked severe combined immunodeficiency (X-SCID) is a combined cellular and humoral immunodeficiency resulting from lack of T and natural killer (NK) lymphocytes and nonfunctional B lymphocytes. Most males with typical X-SCID come to medical attention between three and six months of age. Nearly universal features during the first year of life are failure to thrive, oral/diaper candidiasis, absent tonsils and lymph nodes, recurrent infections, infections with opportunistic organisms such as Pneumocystis, and persistence of infections despite conventional treatment. Additional common features include rashes, diarrhea, cough and congestion, fevers, pneumonia, sepsis, and other severe bacterial infections. Males with atypical X-SCID may have immune dysregulation and autoimmunity associated with rashes, gastrointestinal malabsorption, other autoimmune conditions, and short stature.

Diagnosis/testing. The diagnosis of X-SCID must be made by lymphocyte counts, lymphocyte cell surface staining and enumeration by flow cytometry, lymphocyte functional tests, and molecular genetic testing. A low absolute lymphocyte count compared to age-matched normal infants is usually observed. The number of T cells is usually very low; B cells are generally present but nonfunctional. The number of NK cells is low or absent. IL2RG is the only gene known to be associated with X-SCID. Sequence analysis of the IL2RG coding region detects a mutation in more than 99% of affected individuals. Such testing is available on a clinical basis.

Management. Children with X-SCID require prompt immune reconstitution by bone marrow transplantation (BMT) for survival. BMT is usually performed using HLA-matched bone marrow from a relative or haploidentical parental bone marrow depleted of mature T cells. Interim management during immune reconstitution includes treatment of infections and use of immunoglobulin infusions and antibiotics, particularly prophylaxis against Pneumocystis. Long-term periodic administration of immunoglobulin may be required in those who fail to develop allogeneic, functional B lymphocytes. Gene therapy using autologous bone marrow stem/progenitor cells retrovirally transduced with a therapeutic gene has been successful for immune reconstitution for some individuals, but is only considered for those who are not candidates for BMT or have failed BMT. Prenatal diagnosis with molecular genetic testing ensures that searches can be initiated for bone marrow donors before birth and an affected newborn can be treated immediately by BMT.

Genetic counseling. X-SCID is inherited in an X-linked manner. More than one-half of affected males have no family history of early deaths in maternal male relatives. If the mother of a proband is a carrier, the chance of transmitting the disease-causing mutation in each pregnancy is 50%. Male sibs who inherit the mutation will be affected; female sibs who inherit the mutation will be carriers and will not be affected. Males with X-SCID will pass the disease-causing mutation to all of their daughters and none of their sons. Prenatal testing is possible for pregnancies of women who are carriers for a known IL2RG mutation.

Diagnosis

Clinical Diagnosis

X-linked severe combined immunodeficiency (X-SCID) is a combined cellular and humoral immunodeficiency resulting from lack of T lymphocytes and nonfunctional B lymphocytes. Natural killer (NK) lymphocytes are most often absent as well. X-SCID should be considered in male infants with recurrent or persistent infections that are severe, that do not respond to ordinary treatment, that are caused by opportunistic pathogens, or that cause failure to thrive [Puck 1999, Belmont & Puck 2001].

Testing

The diagnosis of X-SCID must be made by lymphocyte counts, lymphocyte cell surface staining and enumeration by flow cytometry, lymphocyte functional tests, and molecular genetic testing [Buckley et al 1997; Puck, Middelton et al 1997; Puck 1999].

Lymphocyte count. A low absolute lymphocyte count compared to age-matched normal infants is usually observed (see Table 1) [Buckley et al 1997, Myers et al 2002].

  • The number of T cells is usually very low.

  • B cells are generally present, but nonfunctional.

  • The number of NK cells is low or absent.

  • Typical X-SCID is designated TB+NK.

Table 1. Lymphocyte Counts in Infants with X-linked Severe Combined Immunodeficiency

Cell
Type
Lymphocyte CountsControl Values
AverageRange% of Affected IndividualsAverageRange
Total
lymphocytes
<2,000 70%7,300 1 4,000-
13,500 1
5,500 2 >2,000 2
T cells2000-80090-95%5,500>1,800
B cells1,3000->3,0005%800700-1,300
NK cells<100 88%800

Adapted from Puck 1999, Buckley et al 1997, Buckley et al 1999, Conley et al 1990, Stephan et al 1993, and unpublished data. See also Conley & Stiehm 1996.
1. Six months [Altman 1961]
2. Cord blood [Altman 1961]

Lymphocyte functional tests

  • Antibody responses to vaccines and infectious agents are absent.

  • T-cell responses to mitogens are lacking.

Immunoglobulin concentrations

  • Serum concentrations of IgA and IgM are low.

  • IgG is generally normal at birth, but declines as maternally transferred IgG disappears by three months of age.

Thymus. The thymic shadow is absent on chest radiogram.

Molecular Genetic Testing

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Gene. IL2RG is the only gene known to be associated with X-SCID.

Molecular genetic testing: Clinical uses

Molecular genetic testing: Clinical methods

Table 2 summarizes molecular genetic testing for this disorder.

Table 2. Molecular Genetic Testing Used in X-Linked Severe Combined Immunodeficiency

Test MethodMutations DetectedMutation Detection Rate 1 Test Availability
Sequence analysis Missense and nonsense mutations, splice and regulatory regions, insertions>99%Clinical graphic element
Targeted mutation analysis (Southern blot analysis) Large deletions and complex mutations

Interpretation of test results. For issues to consider in interpretation of sequence analysis results, click here.

Clinical Description

Natural History

Typical X-SCID. Affected males appear normal at birth. As transplacentally transferred maternal serum antibody concentrations decline, infants with X-SCID are increasingly prone to infection. Most infants come to medical attention between three and six months of age. Infections that initially appear ordinary such as oral thrush, otitis media, respiratory viral infections (e.g., RSV, parainfluenza 3, adenovirus, influenza), and gastrointestinal diseases resulting in diarrhea may only cause concern when they do not respond to usual medical management.

Nearly universal features during the first year of life are failure to thrive, oral/diaper candidiasis, absent tonsils and lymph nodes, recurrent infections, infections with opportunistic organisms such as Pneumocystis, and persistence of infections. Additional common features include rashes, diarrhea, cough and congestion, fevers, pneumonia, sepsis, and other severe bacterial infections.

Less common features:

  • Disseminated infections (salmonella, varicella, cytomegalovirus, Epstein-Barr virus, herpes simplex virus, BCG, and vaccine strain [live] polio virus)

  • Transplacental transfer of maternal lymphocytes to the infant prenatally or during parturition that causes graft-vs-host disease (GVHD) characterized by erythematous skin rashes, hepatomegaly, and lymphadenopathy [Denianke et al 2001]

  • Recurrent bacterial meningitis

Atypical X-SCID. Less frequently seen are individuals with mutations that result in production of a small amount of gene product or a protein with residual activity. These individuals may have an atypical disease characterized as T+B+NK. These individuals may have immune dysregulation and autoimmunity associated with rashes, splenomegaly, gastrointestinal malabsorption, other autoimmune conditions, and short stature [DiSanto et al 1994; Schmalstieg et al 1995; Puck, unpublished].

Genotype-Phenotype Correlations

Most disease-causing mutations are functionally null. Individuals with rare missense or regulatory mutations may have atypical X-SCID.

Nomenclature

Before the T-cell defect in X-SCID was recognized, X-SCID was included in the designation "Swiss-type agammaglobulinemia." This term is no longer used.

Prevalence

The incidence of X-SCID is unknown, but is estimated to be at least 1/50,000 -100,000 births. All ethnic groups are affected in equal frequency. Because of population structure, X-SCID may account for a larger proportion of individuals with all types of SCID in the United States than in Europe.

Differential Diagnosis

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Severe combined immunodeficiency (SCID) can be classified by the nature of T, B, and NK lymphocyte numbers and function (Table 3) [Buckley 2004]. Presence of each subclass of lymphocytes in most individuals of each genotype is indicated by (+); absence by (–). X-SCID is the most commmon form of SCID. The clinical presentation of X-SCID, JAK3-SCID and IL7RA-SCID is identical. In X-SCID, only males are affected; in JAK3- and IL7R1-SCID, both males and females are affected.

Table 3. Types of SCID

Disease NameGeneLymphocyte PhenotypeInheritanceComments
T B NK
X-SCIDIL2RG +XLRMajority of individuals with SCID
JAK3-SCIDJAK3 AR
IL7RA-SCIDIL7RA ++AR
CD45 deficiencyCD45 +AR
ADA deficiencyADA AR
RAG-deficient SCIDRAG1 +AR
RAG2
SCID AthabascanARTEMIS Athabascan-speaking Native Americans (Navajo, Apache, and others) (10% carrier rate); also other ethnicities

Other X-linked immunodeficiencies includeX-linked agammaglobulinemia, Wiskott-Aldrich syndrome, X-linked hyper-IgM syndrome, X-linked lymphoproliferative disease, NEMO (X-linked ectodermal dysplasia with varying immunodeficiency) (see Incontinentia Pigmenti), IPEX (autoimmunity, polyendocrinopathy, enteropathy), chronic granulomatous disease (CGD), and properdin deficiency.

Human immunodeficiency virus (HIV). Infants with HIV may also have recurrent and opportunistic infections and failure to thrive. They have evidence of HIV virus by p24 antigen testing or PCR testing. In contrast to SCID, T cells are generally present.

Management

Evaluations at Initial Diagnosis to Establish the Extent of Disease

  • History, including family history, growth and development, localized and generalized infectious processes, such as diarrhea, failure to thrive, pneumonia, sepsis, viral and fungal infections

  • CBC and differential count to document absolute lymphocyte count

  • Flow cytometric determination of T-cell, B-cell, NK-cell numbers

  • In vitro mitogenesis assay of patient mononuclear cells stimulated with mitogens (PHA, ConA, PWM) and soluble antigens (Candida antigen, tetanus toxoid)

Treatment of Manifestations

  • Diagnosis of X-SCID demands emergent treatment to provide a functional immune system (see Prevention of Primary Manifestations).

  • Interim management includes treatment of infections and use of immunoglobulin infusions and antibiotics, particularly prophylaxis against jirovecii (formerly Pneumocystis carinii).

Prevention of Primary Manifestations

Bone marrow transplantation (BMT). Prompt immune reconstitution is required for survival of children with X-SCID [Myers et al 2002]. BMT was first successful in 1968 and remains the standard means of immune reconstitution. It is estimated that over 90% of infants with X-SCID can be successfully treated with BMT [Myers et al 2002, Antoine et al 2003]. Although many centers have expertise in performing transplantation in individuals with malignancy, the special issues arising in transplantation for X-SCID require immunodeficiency specialists to be involved for an optimal outcome.

  • HLA-matched bone marrow transplantation from a relative is preferred; however, most individuals lack a matched, related donor.

  • For infants who do not have a matched, related donor, haploidentical parental bone marrow that has been depleted of mature T cells can be used [Buckley et al 1999]. In this technique, the bone marrow is depleted of T cells in order to remove mismatched T cells, which would react against the baby's tissues and cause GVHD.

  • Matched, unrelated donor transplantation of bone marrow or cord blood stem cells has been used in some transplantation centers, but GVHD is a significant problem.

  • In addition, some centers use peripherally harvested hematopoietic stem cells for transplants.

Mismatched T cells would react against the baby's tissues and cause GVHD. Cord blood from normal infants is now being banked; frozen cells can be thawed and used as in other unrelated donor transplants.

The best timing for BMT is immediately after birth because young infants are less likely to have had serious infections or failure to thrive than older infants. Younger infants also have more rapid engraftment, fewer post-transplantation infections, less GVHD, and shorter hospitalizations than those in whom BMT is delayed [Kane et al 2001, Myers et al 2002].

Complications following BMT in some individuals include GVHD, failure to make adequate antibodies requiring long-term immunoglobulin replacement, late loss of T cells, and lymphocyte dysregulation.

The oldest surviving individuals with X-SCID received HLA-matched related BMT and are now in their 30s and healthy.

Administration of immunoglobulin. Long-term periodic administration of immunoglobulin may be required in those who fail to develop allogeneic, functional B lymphocytes.

Gene therapy. Gene therapy performed using autologous bone marrow stem/progenitor cells retrovirally transduced with a therapeutic gene has also been successful in reconstituting the immune system in individuals with X-SCID [Hacein-Bey-Abina et al 2002]; however, the youngest two plus an additional older infant of the first ten infants treated in a French study have developed leukemia as a result of retroviral insertional mutagenesis. Infants with X-SCID in England were also treated successfully with gene therapy with no occurrence of leukemia to date [Gaspar et al 2004]. Two older adolescents did not experience immune reconstitution following attempted gene transfer therapy [Thrasher et al 2005]. Gene therapy is currently only considered for those who are not candidates for BMT or have failed BMT [Gansbacher 2003].

Prevention of Secondary Complications

  • Only CMV-negative, irradiated (1500 to 5000 RADS) blood products should be used.

  • Immunizations should be deferred until after restoration of immunocompetence.

Surveillance

After successful bone marrow transplantation, routine evaluation of affected boys every six to 12 months is appropriate to monitor growth, immune and lung function, and gastrointestinal and dermatologic issues.

Agents/Circumstances to Avoid

  • Individuals with X-SCID should not receive live vaccines.

  • Transfusion of non-irradiated blood products must be avoided.

Testing of Relatives at Risk

In one study, most couples at risk of having an affected pregnancy desired prenatal testing whether or not termination of pregnancy was a consideration [Puck, Middelton et al 1997]. Prenatal testing helped families and professionals prepare for optimal treatment of an affected newborn: bone marrow transplantion centers were chosen, HLA testing of family members and the prenatal sample was carried out, and a search for a marrow donor could be initiated.

Therapies Under Investigation

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mode of Inheritance

X-SCID is inherited in an X-linked manner.

Risk to Family Members

Parents of a proband

Direct DNA testing can often determine the family member in whom the mutation initially arose. Determining the family member in whom a de novo mutation arose is important for determining which branches of the family are at risk for X-SCID.

Sibs of a proband

Offspring of a proband. Males with X-SCID will pass the disease-causing mutation to all of their daughters and none of their sons.

Other family members of the proband. The proband's maternal female relatives may be at risk of being carriers, and their offspring, depending upon their gender, may be at risk of being carriers or of being affected.

Carrier Detection

Carrier testing of at-risk female relatives is available on a clinical basis. See Molecular Genetic Testing section.

Related Genetic Counseling Issues

Family planning. The optimal time for determination of genetic risk, clarification of carrier status, and discussion of the availability of prenatal testing is before pregnancy.

DNA banking. DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. While DNA banking may not be necessary if a disease-causing mutation in the IL2RG gene is identified as causing X-SCID in a family, it may be helpful for other types of SCID. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals. DNA banking is particularly relevant in situations in which the sensitivity of currently available testing is less than 100%. See DNA Banking for a list of laboratories offering this service.

Carrier testing of minors. Carrier testing of at-risk female relatives under the age of 18 years warrants consideration of specific issues, including the minor's experience (or lack of experience) with the disorder, the implications of the test results, the significance that she attributes to them, and the likelihood of her becoming a mother in the near future. The possible benefits of early testing and disclosure must be weighed against the potential harm of loss of autonomy [Fanos, Davis et al 2001; Fanos & Puck 2001]. It is important to assess the minor's ability to understand options and consequences. A minor who can project into the future and has a stable set of values with which to weigh options is the best candidate to participate in determining whether or not to undergo X-SCID carrier testing. A current NIH study is investigating the effects of learning carrier status in females between the ages of 12 and 18 years who decide to undergo X-SCID carrier testing.

Prenatal Testing

Molecular genetic testing. Prenatal testing is possible for pregnancies of women who are carriers for X-SCID [Puck, Middelton et al 1997]. The usual procedure is to perform chromosome analysis for sex determination on fetal cells obtained by chorionic villus sampling (CVS) at about 10-12 weeks' gestation or by amniocentesis at about 15-18 weeks' gestation. If the karyotype is 46,XY and if the disease-causing IL2RG mutation has been identified in a family member, DNA from fetal cells can be analyzed for the known disease-causing mutation.

Note: Gestational age is expressed as menstrual weeks calculated either from the first day of the last normal menstrual period or by ultrasound measurements.

Percutaneous umbilical blood sampling (PUBS). When the family-specific mutation is not known, fetal blood sampling is considered in some centers. Fetal blood is analyzed for lymphoctyopenia, low numbers of T cells, and poor T-cell blastogenic responses to mitogens, all of which can be definitively demonstrated in affected fetuses by 17 weeks of gestation; however, caution is necessary as maternal blood contamination can make test results look falsely normal. Involvement of experienced high-risk perinatologists, genetics experts, and immunologists is advised.

Molecular Genetics

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Table A. Molecular Genetics of X-Linked Severe Combined Immunodeficiency

Gene SymbolChromosomal LocusProtein Name
IL2RGXq13Cytokine receptor common gamma chain

Data are compiled from the following standard references: Gene symbol from HUGO; chromosomal locus, locus name, critical region, complementation group from OMIM; protein name from Swiss-Prot.

Table B. OMIM Entries for X-Linked Severe Combined Immunodeficiency

300400 SEVERE COMBINED IMMUNODEFICIENCY, X-LINKED; SCIDX1
308380 INTERLEUKIN 2 RECEPTOR, GAMMA; IL2RG

Table C. Genomic Databases for X-Linked Severe Combined Immunodeficiency

Gene SymbolLocus SpecificEntrez GeneHGMD
IL2RGIL2RG3561 (MIM No. 308380)IL2RG

For a description of the genomic databases listed, click here.

Normal allelic variants: The gene spans 4.5 kb of genomic DNA. The coding sequence of 1,124 nucleotides is divided into eight exons. There are no common normal allelic variants.

Pathologic allelic variants: Over 300 mutations have been identified spanning all eight exons of the gene. They are primarily single-nucleotide changes or changes of a few nucleotides, small insertions, deletions, and splice defects. Mutation hot spots in the IL2RG are reported [Puck 1996; Puck, Pepper et al 1997; Puck 1997]. (For more information, see Genomic Databases table above.)

Normal gene product: The normal gene product is the common gamma chain, or gamma-c, which is a transmembrane protein in the cytokine receptor gene superfamily. It is a component of multiple cytokine receptors on the surface of lymphocytes and other hematopoietic cells, including the receptors for IL-2, -4, -7, -9, -15, and 21.

Abnormal gene product: Although over two-thirds of mutations result in lack of protein expression, truncated gamma-c proteins or gamma-c proteins bearing amino acid substitutions, insertions, or deletions have been described and are nonfunctional.

Resources

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

National Library of Medicine Genetics Home Reference
X-linked severe combined immunodeficiency

Immune Deficiency Foundation
40 W Chesapeake Ave; Suite 308
Towson, MD 21204
Phone: 800-296-4433; 410-321-6647
Fax: 410-321-9165
Email: idf@primaryimmune.org
www.primaryimmune.org

International Patient Organisation for Patients with Primary Immunodeficiencies
Firside Main Road
Downderry
Cornwall PL11 3LE
United Kingdom
Email: david@pia.org.uk
http://ipopi.org/

Jeffrey Modell Foundation/National Primary Immunodeficiency Resource Center
747 Third Avenue, 34A
New York, NY 10017
Phone: 800-533-3844; 212-819-0200
Fax: 212-764-4180
Email: info@jmfworld.org
www.info4pi.org

Primary Immunodeficiency Diseases Registry (PIDR)
Phone: 800-296-4433
Primary Immunodeficiency Registry

References

Medical Genetic Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page. graphic element

Literature Cited

Altman PL. Blood leukocyte values: man. In: Dittmer DS (ed) Blood and Other Body Fluids. Federation of American Societies for Experimental Biology, Washington, DC, PP 125-6. 1961
Antoine C, Muller S, Cant A, Cavazzana-Calvo M, Veys P, Vossen J, Fasth A, Heilmann C, Wulffraat N, Seger R, Blanche S, Friedrich W, Abinun M, Davies G, Bredius R, Schulz A, Landais P, Fischer A. Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: report of the European experience 1968-99. Lancet. 2003; 361: 55360. [PubMed]
Belmont JW, Puck JM. T cell and combined immunodeficiency syndromes. In: Scriver DR, Beaudet AL, Sly WS (eds) The Metabolic and Molecular Bases of Inherited Disease, 8 ed. McGraw-Hill, New York, pp 4751-84. 2001
Buckley RH. Molecular defects in human severe combined immunodeficiency and approaches to immune reconstitution. Annu Rev Immunol. 2004; 22: 62555. [PubMed]
Buckley RH, Schiff RI, Schiff SE, Markert ML, Williams LW, Harville TO, Roberts JL, Puck JM. Human severe combined immunodeficiency: genetic, phenotypic, and functional diversity in one hundred eight infants. J Pediatr. 1997; 130: 37887. [PubMed]
Buckley RH, Schiff SE, Schiff RI, Markert L, Williams LW, Roberts JL, Myers LA, Ward FE. Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency. N Engl J Med. 1999; 340: 50816. [PubMed]
Conley ME, Buckley RH, Hong R, Guerra-Hanson C, Roifman CM, Brochstein JA, Pahwa S, Puck JM. X-linked severe combined immunodeficiency. Diagnosis in males with sporadic severe combined immunodeficiency and clarification of clinical findings. J Clin Invest. 1990; 85: 154854. [PubMed]
Conley ME, Lavoie A, Briggs C, Brown P, Guerra C, Puck JM. Nonrandom X chromosome inactivation in B cells from carriers of X chromosome-linked severe combined immunodeficiency. Proc Natl Acad Sci U S A. 1988; 85: 30904. [PubMed]
Conley ME, Stiehm ER. Immunodeficiency Disorders. In: Stiehm ER (ed) Immunologic Disorders in Infants and Children. WB Saunders and Company, pp 216-17. 1996
Denianke KS, Frieden IJ, Cowan MJ, Williams ML, McCalmont TH. Cutaneous manifestations of maternal engraftment in patients with severe combined immunodeficiency: a clinicopathologic study. Bone Marrow Transplant. 2001; 28: 22733. [PubMed]
DiSanto JP, Rieux-Laucat F, Dautry-Varsat A, Fischer A, de Saint Basile G. Defective human interleukin 2 receptor gamma chain in an atypical X chromosome-linked severe combined immunodeficiency with peripheral T cells. Proc Natl Acad Sci U S A. 1994; 91: 946670. [PubMed]
Fanos JH, Davis J, Puck JM. Sib understanding of genetics and attitudes toward carrier testing for X-linked severe combined immunodeficiency. Am J Med Genet. 2001; 98: 4656. [PubMed]
Fanos JH, Puck JM. Family pictures: growing up with a brother with X-linked severe combined immunodeficiency. Am J Med Genet. 2001; 98: 5763. [PubMed]
Gansbacher B. Report of a second serious adverse event in a clinical trial of gene therapy for X-linked severe combined immune deficiency (X-SCID). Position of the European Society of Gene Therapy (ESGT). J Gene Med. 2003; 5: 2612. [PubMed]
Gaspar HB, Parsley KL, Howe S, King D, Gilmour KC, Sinclair J, Brouns G, Schmidt M, Von Kalle C, Barington T, Jakobsen MA, Christensen HO, Al Ghonaium A, White HN, Smith JL, Levinsky RJ, Ali RR, Kinnon C, Thrasher AJ. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet. 2004; 364: 21817. [PubMed]
Hacein-Bey-Abina S, Le Deist F, Carlier F, Bouneaud C, Hue C, De Villartay JP, Thrasher AJ, Wulffraat N, Sorensen R, Dupuis-Girod S, Fischer A, Davies EG, Kuis W, Leiva L, Cavazzana-Calvo M. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med. 2002; 346: 118593. [PubMed]
Kane L, Gennery AR, Crooks BN, Flood TJ, Abinun M, Cant AJ. Neonatal bone marrow transplantation for severe combined immunodeficiency. Arch Dis Child Fetal Neonatal Ed. 2001; 85: F1103. [PubMed]
Myers LA, Patel DD, Puck JM, Buckley RH. Hematopoietic stem cell transplantation for severe combined immunodeficiency in the neonatal period leads to superior thymic output and improved survival. Blood. 2002; 99: 8728. [PubMed]
Noguchi M, Yi H, Rosenblatt HM, Filipovich AH, Adelstein S, Modi WS, McBride OW, Leonard WJ. Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. Cell. 1993; 73: 14757. [PubMed]
O'Marcaigh AS, Puck JM, Pepper AE, De Santes K, Cowan MJ. Maternal mosaicism for a novel interleukin-2 receptor gamma-chain mutation causing X-linked severe combined immunodeficiency in a Navajo kindred. J Clin Immunol. 1997; 17: 2933. [PubMed]
Puck JM. IL2RGbase: a database of gamma c-chain defects causing human X-SCID. Immunol Today. 1996; 17: 50711. [PubMed]
Puck JM. Primary immunodeficiency diseases. JAMA. 1997; 278: 183541. [PubMed]
Puck JM. X-linked severe combined immunodeficiency. In: Ochs H, Smith CIE, Puck JM (eds) Primary Immunodeficiency Diseases, a Molecular and Genetic Approach. Oxford University Press, New York, pp 99-110. 1999
Puck JM, Deschenes SM, Porter JC, Dutra AS, Brown CJ, Willard HF, Henthorn PS. The interleukin-2 receptor gamma chain maps to Xq13.1 and is mutated in X-linked severe combined immunodeficiency, SCIDX1. Hum Mol Genet. 1993; 2: 1099104. [PubMed]
Puck JM, Middelton L, Pepper AE. Carrier and prenatal diagnosis of X-linked severe combined immunodeficiency: mutation detection methods and utilization. Hum Genet. 1997; 99: 62833. [PubMed]
Puck JM, Nussbaum RL, Conley ME. Carrier detection in X-linked severe combined immunodeficiency based on patterns of X chromosome inactivation. J Clin Invest. 1987; 79: 1395400. [PubMed]
Puck JM, Pepper AE, Henthorn PS, Candotti F, Isakov J, Whitwam T, Conley ME, Fischer RE, Rosenblatt HM, Small TN, Buckley RH. Mutation analysis of IL2RG in human X-linked severe combined immunodeficiency. Blood. 1997; 89: 196877. [PubMed]
Schmalstieg FC, Leonard WJ, Noguchi M, Berg M, Rudloff HE, Denney RM, Dave SK, Brooks EG, Goldman AS. Missense mutation in exon 7 of the common gamma chain gene causes a moderate form of X-linked combined immunodeficiency. J Clin Invest. 1995; 95: 116973. [PubMed]
Stephan JL, Vlekova V, Le Deist F, Blanche S, Donadieu J, De Saint-Basile G, Durandy A, Griscelli C, Fischer A. Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients. J Pediatr. 1993; 123: 56472. [PubMed]
Thrasher AJ, Hacein-Bey-Abina S, Gaspar HB, Blanche S, Davies EG, Parsley K, Gilmour K, King D, Howe S, Sinclair J, Hue C, Carlier F, von Kalle C, de Saint Basile G, le Deist F, Fischer A, Cavazzana-Calvo M. Failure of SCID-X1 gene therapy in older patients. Blood. 2005; 105: 42557. [PubMed]
Wengler GS, Allen RC, Parolini O, Smith H, Conley ME. Nonrandom X chromosome inactivation in natural killer cells from obligate carriers of X-linked severe combined immunodeficiency. J Immunol. 1993; 150: 7004. [PubMed]

Published Statements and Policies Regarding Genetic Testing

No specific guidelines regarding genetic testing for this disorder have been developed.

Chapter Notes

Revision History

  • 12 December 2005 (me) Comprehensive update posted to live Web site

  • 26 August 2003 (me) Review posted to live Web site

  • 23 April 2003 (jd) Original submission

Next
GeneReviews2009
(navigation arrows) Go to previous chapter Go to next chapter Go to top of this page Go to bottom of this page Go to Table of Contents