HRSA - U.S Department of Health and Human Services, Health Resources and Service Administration HHS
Home
Questions
Order Publications
 
Maternal & Child Health

Advisory Committee on Heritable Disorders in Newborns and Children

 

Committee Report
Fifteenth Meeting
October 1-2, 2008

Adobe Acrobat PDF Printer-friendly Committee Report (111 KB)

EVIDENCE REVIEW:  Severe Combined Immunodeficiency (SCID)

 

On this page:

i. Abbreviations Used 
I. Overview 
II. Rationale for Review
III. Objectives of Review
IV. Main Questions
V. Conceptual Framework
VI. Methods
VII. Results
VIII. Summary
IX. References         

 

i. ABBREVIATIONS USED

ACHDNC Advisory Committee on Heritable Disorders in Newborns and Children
ACTB  Gene symbol for beta-actin
ADA-deficiency Adenosine deaminase deficiency
BMT Bone marrow transplant
B+ SCID Subgroup of severe combined immunodeficiency in which B lymphocytes are present
B- SCID Subgroup of severe combined immunodeficiency in which B lymphocytes are absent
CD Antigens Cluster of Differentiation, nomenclature of leucocyte molecules
CI Confidence interval
CIBMTR Center for International Blood and Marrow Transplant Research
CMV Cytomegalovirus
DNA Deoxyribonucleic acid
ELISA Enzyme-Linked ImmunoSorbent Assay
ERG Evidence review group
ERT Enzyme replacement therapy
G-CSF Granulocyte-colony stimulating factor
GvHD Graft versus host disease
HLA Human leukocyte antigen
HSCT Hematopoietic stem cell transplant
IgA Immunoglobulin A
IgG  Immunoglobulin G
IgM Immunoglobulin M
IL-7 Interleukin 7
IVIG Intravenous immunoglobulin
JAK Janus kinase
MeSH National Library of Medicine medical subject heading
MMRD Mismatched Related Donor
MUD Matched Unrelated Donor
NIAID National Institute of Allergy and Infectious Diseases
NK Natural killer
PEG Polyethylene Glycol
PHA Phytohemagglutinin
RID Related Identical
SCID Severe Combined Immunodeficiency
SCIDA Severe Combined Immunodeficiency, Athabaskan-Type
TREC T-cell receptor excision circles
USIDNET United States Immunodeficiency Network
XSCID X-Linked Severe Combined Immunodeficiency
gammac  Common cytokine receptor gamma chain 


I.  OVERVIEW: 
Severe Combined Immunodeficiency (SCID) is a group of disorders characterized by the absence of both humoral and cellular immunity.  Reported incidence is approximately 1/100,000 (Chan, Puck 2005, McGhee et al. 2005). Currently at least 15 genes are known to cause SCID when mutated (Puck, SCID Newborn Screening Working Group 2007).  Although disease presentation varies (Buckley et al. 1997), as protection from maternal antibodies wanes during the first months of life, infants with SCID develop infections due to both common and opportunistic pathogens (Buckley et al. 1997).  Treatment and prevention of infections can prolong life but are not curative actions (Buckley et al. 1999).

Early receipt of hematopoietic stem cell transplant (HSCT), prior to the onset of severe infections, offers the best chance of cure (Buckley et al. 1999).  Although family history leads to early detection of some infants, the majority of infants with SCID are not detected until they develop clinical symptoms such as recurrent infections, failure to thrive and infection with opportunistic organisms (Buckley et al. 1997).  The apparent value of early HSCT has led to a search for methods for pre-symptomatic identification of infants with SCID, including possible population-based newborn screening.

II. RATIONALE FOR REVIEW
The Advisory Committee on Heritable Disorders in Newborns and Children has directed the Evidence Review Group to produce this report for the nominated condition of SCID.  SCID has been nominated for the following reasons:

  1. Without treatment SCID leads to death in early childhood. 
  2. Earlier treatment, particularly before the onset of lung infection, decreases the mortality and morbidity associated with SCID and with HSCT for SCID.
  3. Methods to screen infants for SCID using quantitative PCR for T-cell receptor excision circles (TREC) have been developed.

III. OBJECTIVES OF REVIEW

 

The objective of this review is to provide information to the Advisory Committee on Heritable Disorders in Newborns and Children (ACHDNC) to guide recommendations regarding screening newborns for SCID.  Specifically, the Evidence Review Group’s (ERG) goal was to summarize the evidence available from published studies, as well as critical unpublished data available from key investigators in the field.

MAIN QUESTIONS

 

We sought to answer the following questions, with a particular emphasis on the questions related to screening and the benefits of early treatment.

  1. What is the natural history of SCID and are there clinically important phenotypic or genotypic variations? 
  2. What is the prevalence of SCID and its variations?
  3. What methods exist to screen newborns for SCID?  How accurate are those methods?  What are their sensitivity and specificity?  What methods exist to diagnose individuals with positive screens?
  4. What benefit does treatment, particularly pre-symptomatic, confer?
  5. What are the potential harms or risks associated with screening, diagnosis or treatment?
  6. What costs are associated with screening, diagnosis, treatment and the failure to diagnose in the newborn period?

By answering these questions we hoped to provide the ACHDNC with answers to broader questions related to SCID screening.  Specifically:

  1. Do current screening tests effectively and efficiently identify cases of SCID?
  2. Does pre-symptomatic or early symptomatic intervention in newborns or infants with SCID improve health outcomes?  In other words, does early identification lead to better outcomes?
  3. What is the cost-effectiveness of newborn screening for SCID?
  4. What critical evidence appears lacking that may inform screening recommendations for SCID?

V.  CONCEPTUAL FRAMEWORK

The conceptual framework illustrated below outlines the salient factors when considering newborn screening for any disorder, SCID in the case of this review.   Children will develop SCID regardless of whether or not they are screened.  The key decision points, therefore, are whether to screen newborns and what treatment to pursue for affected children identified.  At either decision point, children may experience benefits from the decision but also are at risk of adverse effects, including false positive or false negative screening results and significant treatment side-effects.  The combination of the baseline risk for SCID and the effects of the screening and treatment decisions lead to a state of health. 

Figure 1- Conceptual Framework

figure 1

VI. METHODS

This evidence review provides information from two sources: a systematic literature review and interviews with experts in the field of SCID.

A.   Literature Review
We searched MEDLINE for all relevant studies published over the 20 year period from January 1988 to October 2008.  We completed searches combining the National Library of Medicine Medical Subject Heading (MeSH) “severe combined immunodeficiency” with each of the following MeSH terms: epidemiology, incidence, prevalence, disease progression, mass screening, neonatal screening, genetic screening, diagnosis, and therapeutics.  In order to capture articles which have not yet been assigned MeSH terms, we also searched the following keywords within the OVID In-Process and Other Non-Indexed Citations database: severe combined immunodeficiency, severe immune deficiency, adenosine deaminase deficiency.  We applied human studies and English language limitations to all searches.  This search strategy resulted in a total of 725 articles.

All abstracts were reviewed by at least 2 readers (EL, JD, JP).  Articles which were non-human data, reviews, editorials or other opinion pieces, case-series of <4 patients, only contained adult subjects or did not address one or more of the key questions were eliminated.  Disagreements were resolved through discussion with emphasis on inclusion of any potentially useful data.  After this process 60 articles remained which were reviewed in detail.

Each article was evaluated, using standardized tools, for the quality of the study design (NHS Centre for Reviews and Dissemination, March 2001) and the quality of the evidence, as it relates to the category of evidence (Pandor et al. 2004, Pollitt et al. 1997).  A given article received only one rating per reader for study design, but may have received multiple quality evaluations for the type of evidence.  For example, a study that discusses prevalence and natural history would be evaluated for the quality of the evidence in each of those domains.

Data were abstracted from the articles by 2 reviewers (EL, SV).  A subset of the papers, approximately 20%, was abstracted by both reviewers in order to check for agreement.  There were no significant differences in the data extracted by the reviewers.

B.  Interviews with Experts
The ERG and the ACHDNC recognize that in a rapidly developing field, such as newborn screening for SCID, there may be crucial evidence that is either not yet ready for publication or has not been published for other reasons.  As such, using our literature search and discussions with content experts, we identified a list of key investigators.  This list was discussed among the members of the ERG, and additional names were added based on personal knowledge of the field by members of the ERG.  This list (Table 1) was approved by the ERG and the experts were contacted via e-mail.  Reminder e-mails were sent to non-responders approximately 2 weeks after the initial e-mail.

Experts were sent a letter (appendix) explaining the purpose of the review, a conflict of interest form (appendix) and an open-ended survey (appendix).  After receipt of the conflict of interest form and survey, phone interviews were scheduled between experts and members of the ERG (EL, AK, JP), when clarification of the written survey was needed.  The semi-structured phone interviews clarified information provided in the written survey.  Information from the surveys and interviews is explicitly provided in this report when such information was not available in the published literature.

Table 1- List of experts contacted and degree of participation

Name

Title

Completed Written Survey

Telephone
Interview

Mei Baker

Assistant Professor, Department of Pediatrics, School of Medicine and Public Health, Science Advisor, Newborn Screening Program, Wisconsin State Laboratory of Hygiene
University of Wisconsin-Madison

check mark

check mark

Tony (Francisco) Bonilla

Program Director, Clinical Immunology Assistant in Medicine, Assistant Professor, Harvard Medical School and Children’s Hospital, Boston, Mass

check mark*

 

Rebecca Buckley

Professor of Pediatrics, Professor of Immunology,
Duke University Medical Center, Durham, North Carolina

check mark

check mark

Anne Comeau

Deputy Director New England Newborn Screening Program University of Massachusetts Medical School

check mark

check mark

Lisa Filipovich

Medical Director, Hematology/Oncology Diagnostic Laboratory, Division of Hematology/ Oncology, Cincinnati Children's Hospital Medical Center

 

 

Alain Fischer

Professor of Pediatric Immunology and Head of the Pediatric Immunology Department and the INSERM Research Unit

 

 

Alan P. Knutsen

Professor of Pediatrics, Allergy and Immunology, Director of Pediatric Clinical Immunology Laboratory, Department of Pathology, St Louis University Health Sciences Center

 

 

Ronald Laessig

Professor of Population Health Sciences and Pathology at University of Wisconsin

 

 

Edward McCabe

Clinical Biochemical Genetics, Clinical Genetics, Professor and Executive Chair of the UCLA Department of Pediatrics, and Physician-in-Chief of the Mattel Children’s Hospital at UCLA

check mark**

 

Sean McGhee

Assistant Clinical Professor of Pediatrics. Division of Pediatric Immunology, Department of Pediatrics,
David Geffen School of Medicine at UCLA, Los Angeles, California

check mark

 

Luigi Notarangelo

Director, Research and Molecular Diagnosis Program on Primary Immunodeficiencies, Division of Immunology, Children's Hospital Boston, Mass
Professor of Pediatrics and Pathology, Harvard Medical School

check mark*

check mark

Hans Ochs

Professor of Pediatrics and Immunology at the University of Washington

 

 

Sung-Yun Pai

Assistant Professor in Pediatrics, Pediatric Hematology/Oncology, Harvard Medical School and Children’s Hospital, Boston, Mass

check mark*

 

Ken Pass

Senior Research Scientist, Molecular Medicine, Wadsworth Center, New York State Department of Health, Albany, New York

check mark

 

Jennifer Puck

Professor, Department of Pediatrics and Institute for Human Genetics University of California, San Francisco

check mark

check mark

Robert Vogt

Centers for Disease Control and Prevention, Newborn Screening Quality Assurance Program, Atlanta, Georgia

check mark

 

*Responded as a group with one combined survey
**Dr. McCabe declined to answer and referred us to Dr. McGhee

VII. RESULTS
Case definition:  For the purpose of this review, Severe Combined Immunodeficiency is defined based on the definition for the PubMed medical subheading.  SCID is a “group of rare congenital disorders characterized by impairment of both humoral and cell-mediated immunity, leukopenia, and low or absent antibody levels. It is inherited as an X-linked or autosomal recessive defect.” (U.S. National Library of Medicine and the National Institutes of Health, 2008)   Children with SCID universally have extremely low or absent T-cells and may or may not have B-cells.  We have included some specific sub-types such as adenosine deaminase deficiency (ADA-deficiency), reticular dysgeneis and Omenn syndrome in the definition of SCID because they are characterized by absence of T-cells, but we recognize that not some groups consider these disorders distinct from SCID (WHO Scientific Group, 1995).

Table 2- Study design for abstracted articles

Study Design

Number of papers

Experimental intervention

0

Cohort study

11

Case-control study

8

Case series total

38

blank

Sample size = 10

11

blank

Sample size 11 to 50

18

blank

Sample size = 51

9

Economic Evaluation

1

Other design

2*

Total

60

*Epidemiologic studies using retrospective record review (Jones et al. 1991) and telephone survey (Boyle & Buckley, 2007)

 

*Epidemiologic studies using retrospective record review (Jones et al. 1991) and telephone survey (Boyle & Buckley, 2007)

Evidence review:  The remainder of this section of the report is dedicated to presentation of the evidence.  Data is presented by key questions.  A table indicating the quality of the evidence for that key question leads the section, followed by data from the literature review.  Finally, any additional information learned from surveying experts is provided at the end of each section.

    1. What is the natural history of SCID and are there clinically important phenotypic or genotypic variations?

 


Table 3- Quality Assessment of abstracted literature pertaining to Natural History
                       


Genotype/Phenotype Correlation

12

Data from retrospective screening studies in U.S. or similar population.

0

Data from systematic studies other than whole population screening.

5

Estimated from the known clinical features of the condition as described for individual cases or short series.

7

 

 

Incidence (cases per 100,000), average within the U.S.

4

Data obtained from whole-population screening or comprehensive national surveys of clinically detected cases.

1

Ia.  As in I but more limited in geographical coverage or methodology.

2

Extrapolated from class I data for non-U.S. populations.

0

Estimated from number of cases clinically diagnosed in U.S.

1

Adapted from Pandor et al. 2004, Pollitt et al. 1997

The term SCID refers to a group of slightly heterogeneous disorders which are all related by the absence of T-cells in affected individuals.  Some subtypes are missing other lymphocyte subsets, such as B-cells or NK-cells, and others have associated non-immune manifestations such as neurocognitive deficits, hearing loss and skeletal abnormalities.
We describe below evidence related to the characteristic presentations of SCID, the natural history (without treatment), subtypes of SCID, and genotype/phenotype data.  The main findings are: 1) with the exception of children diagnosed early in life, typically through prenatal testing initiated because of family history, most children are diagnosed after recurrent pulmonary infections or infections with opportunistic organisms; 2) this is true of all SCID subtypes, although the exact timing may vary; and 3) without treatment of the underlying immunodeficiency, children with SCID die in early childhood from infection. 

General findings regarding natural history and associated clinical conditions -Systematic literature review
Several studies provide descriptive data regarding symptomatic presentation of children with SCID, almost all of which highlighted pulmonary and opportunistic infections, leading to early childhood death, as the key complications of untreated SCID.
A chart review (Deerojanawong et al. 1997) found that all children who did not receive prenatal diagnoses of SCID (13/15) had pulmonary symptoms at presentation.  Of 9 that died 5 were due to pulmonary disease.  Moreover, pulmonary infections, most commonly Pneumocystis jirovevi pneumonia (PJP), were the most common presenting sign of SCID, occurring in 10/15 children, at a median age of 4 months.  Another case series (Stocks et al. 1999) found 14/18 of the children, over a 20-year time period, had otolaryngological signs or symptoms (including congestion, URI, thrush, oral ulcers, cervical adenopathy, otitis media; mastoiditis) prior to diagnosis with SCID.
An institutional case-series (Stephan et al. 1993) of 117 patients treated for SCID between 1970 and 1992, found the median age at diagnosis, which did not change over the time period, was 4.6 months with an average of 2 months delay between clinical manifestations and diagnosis.  The primary clinical manifestations included oral candida, skin erythema, diarrhea with growth impairment and interstitial pneumonitis.   
Hague et al (Hague et al. 1994) found that, among 32 children with no family history of SCID, the median age of diagnosis was 7 months, despite the median age of symptom onset being 5 weeks.  In this group the median age of first hospital admission was 4 months with 22/32 presenting with respiratory infection, 9/32 with vomiting and diarrhea, 8/32 with candidal infection and 6/32 with failure to thrive. 

Two papers discussed physical signs or symptoms that may occur with high prevalence in children with SCID but are not necessarily presenting signs of the disease, including  gastroesophageal reflux (GER) (Boeck, Buckley & Schiff 1997)   and  rashes, particularly due to engraftment of maternal T lymphocytes (Denianke et al. 2001).

Table 4 - Evaluated literature pertaining to natural history of SCID (alphabetical by author)

Study (Author, date)

Population:
Size and age by subgroups

 

Study type

Key findings

Bertrand et al. 1999

178 children:
B+ group: 122,  mean age at BMT 7 months
B- group: 56, mean age at BMT 6.5 months

Cohort study

  • Deaths due primarily to infection.
  • B- SCID had higher rates of both early and late death

Bertrand et al. 2002

10 children: BMT at age 0.5 to 10 months

Case series

  • Children with reticular dysgenesis.
  • Presented with infection in first days of life.  

Buckley et al. 1997

108 children: Diagnosis at Age birth -21 months

Case series

  • 67 (76%) of the 88 families were white, 14 (16%) were black, and 7 (8%) Hispanic.
  • Abnormal serum immunoglobulin concentrations in all types of SCID.
  • Mean IgA levels were lowest in gammac-deficient, Jak3-deficient and unknown types of SCID.
  • Mean IgM concentrations were lowest in ADA, gammac and Jak3 deficient patients. 
  • IgE was normal in all but 2 patients.

Buckley et al. 1999

89 children:
22 were <3.5 months at transplant
67 were greater than or equal to 3.5 months (0-21 months at time of diagnosis) at transplant

Case series

  • Mean number of T/B/NK cells varies by genotype; all genotypes had normal average in vitro mitogen response after transplant.
  • Poor B cell function with 45 kids requiring IVIG.
  • NK activity low in gammac-chain and JAK3 deficiency, otherwise normal.

Cavazzana-Calvo et al. 2007

31 children:
aged 1-42 months at HSCT, now 10-27 years later

 

Case series

  • 26/31 had T-cell count in normal range for age.
  • 30 had normal mitogen induced T-cell proliferation.
  • 18/31 had normal TREC counts.
  • Presence of NK cells not correlated with TREC.
  • gammac deficiency may be associated with TREC detection.
  • Presence of TREC/naïve T-cells associated with significantly higher t-cell counts and better t-cell mitogen proliferation.

 

Gomez et al. 1995

9 children: Aged
0.5-5 months (mean 2.8) and had BMT for Omenn Syndrome between 1981 and 1989

Case series

  • Engraftment occurred in 4/5 HLA identical transplants and 3/4 non-identical.
  • Full chimerism occurred in all but one patient of those that engrafted.
  • One patient died of CMV 50 days post-transplant, 3 developed interstitial pneumonia.
  • Clinical manifestations of Omenn syndrome disappeared within days of BMT (likely due to pre-treatment with chemo.)
  • All survivors except one (who has chronic GVHD) have normal growth rates.

Laffort et al. 2004

41 patients, at least 10 years after HSCT for SCID

 

Case-control study

  • 9/41 developed “extensive chronic” human papilloma virus.
  • 4 had lesions typical of a rare genodermatosis.
  • All with human papilloma virus had gammac/Adenylate kinase- 3 (AK-3) deficiency, though only 9/18 with gammac/JAK3 developed human papilloma virus.
  • gammac /AK-3 with or without human papilloma virus showed lower NK counts than patients with other SCID subtypes.

Monafo et al. 1992

4 children (2 sets of siblings) aged: 20 months, 4 months, 11 months, 24 months at the time of BMT

Case series

  • One child did not present until 1 year of age, with a previously undescribed phenotype.
  • Presenting clinical signs of SCID including diarrhea, poor weight gain, oral ulcers, and PJP pneumonia but normal lymphocyte counts. 
  • CD8 cells were virtually absent and mononuclear cells did not proliferate normally in vitro. 

O'Marcaigh et al. 2001

18 children:
Birth to 3 months at diagnosis; mean of 17.5 months at first transplant

Case series

  • Oral/genital ulcers associated with SCIDA. 
  • SCIDA occurs with relatively high incidence among Athabaskan-speaking Native Americans.

Roberts et al. 2004

170 total patients: 10 with JAK3 deficiency and 160 with other types of SCID

Case series

  • All patients had abnormal B cell JAK3 dependent interleukin 2 induced signal transducer and activator of transcription 5.phosphorylation.
  • 9/10 patients are alive and well 4 to 18 years after stem cell transplant.

Rogers et al. 2001

22 children: aged 1-51 months at transplant; 0.9-18 years at follow-up;
11 ADA-deficient, 11 other SCID types

 

Cohort study

  • No significant difference in full scale IQ between groups (both were below-average), ADA-deficient 2 standard deviations below zero.
  • Significant behavioral differences in ADA-deficient SCID children: more hyperactivity, higher scores for dysfunction in social, emotional and behavioral domains.
  • Abnormal behavior more evident in older children.
  • Gross neurologic evaluation of motor function showed no abnormalities in ADA-deficient group.

 

Genotype specific findings –Systematic literature review
Although as many as 15 different genes (Puck, SCID Newborn Screening Working Group 2007) are associated with SCID, the known phenotypic variation is less extensive.  There is some phenotypic variation, as outlined below, in immunoglobulin levels, lymphocyte counts and lymphocyte function.  More striking, however, a few phenotypes appear to have distinct characteristics at presentation.  Specifically we found evidence supporting very early infections in children with reticular dysgenesis (Bertrand et al. 2002), neurologic symptoms in children with ADA-deficiency (Stephan et al. 1993, Honig et al. 2007), and oral/genital ulcers associated with a distinct form of SCID known as SCIDA occurring among Athabaskan-speaking Native Americans (O'Marcaigh et al. 2001).

A case-series (Buckley et al. 1997) of 108 infants examined the relationship between genotype and immune functional status.  These patients ranged from birth to 21 months at diagnosis and children with less severe forms of combined immunodeficiency, such as Omenn syndrome, were excluded.  The distribution of genotypes from this series is shown below in table 5.

Table 5- Relative frequencies of different SCID genotypes from Buckley et al. 1997

 

SCID Infants* (n=108)

SCID families* (n=88)

Genotype

Number

Percent

Number

Percent

gammac deficiency

49

45.4

37

42

ADA-deficiency

16

14.8

13

15

Jak3 deficiency

8

7.4

5

6

Autosomal Recessive
(not ADA or Jak3 deficiency)

21

19.4

19

22

Reticular Dysgenesis

1

0.9

1

<1

Cartilage-hair hypoplasia

1

0.9

1

<1

Unknown

12

11.1

12

14

*table depicts genotype according to the 108 individual patients in the first column, and the 88 families of origin in the second column where siblings count as one unit

The authors found abnormal serum immunoglobulin concentrations in all types of SCID.  Mean IgA levels were lowest in gammac-deficient, Jak3-deficient and unknown types of SCID.  Mean IgM concentrations were lowest in ADA, gammac and Jak3 deficient patients.  IgE was normal in all but 2 patients.
Lymphopenia was present in all categories of SCID but more severe in patients with ADA-deficient SCID.  5 infants had elevated lymphocytes, of which 4 were due to the presence of maternal cells.  Lymphocyte subpopulations varied between types of SCID.  ADA-deficient children had the lowest mean number of B-cells and gammac and Jak3 the highest.  Mean number of NK cells was lowest in ADA, gammac, and Jak3 deficient patients.  Lymphocyte response to mitogens was diminished in all patients.

B. What is the prevalence of SCID and its variations?

Most methods of determining the prevalence and incidence of a disorder depend on case ascertainment.  As in other disorders with high risk of early death, the incidence of SCID may be higher than measured via case ascertainment because some patients may die prior to having a definitive diagnosis.

Incidence and Prevalence-Systematic literature review
In a study with a primary focus on SCID screening methods, Chan and Puck (Chan, Puck 2005) estimated the annual incidence as a minimum of 1/105,000 births.  This calculation was based on assuming an annual US birthrate of 4,000,000 that all children with XSCID in the U.S. had blood samples sent to the authors’ laboratory for mutation detection (19 in one year) and that XSCID represents approximately half of all cases of SCID.  A study (Stephan et al. 1993) of French children with SCID estimated a minimum incidence of 1/100,000 births.  This estimate is based on the number of French children referred to specialized units over a 5 year time period.
Some US populations, namely the Navajo, have a higher prevalence of SCID because of a founder mutation in the Artemis gene.  The death records (Jones et al. 1991) of all Native American children in Arizona who died between 1969 and 1978 and Navajo children who died between 1969 and 1982 in Arizona and New Mexico were reviewed.  Subsequently the authors reviewed the hospital records of children who died from SCID, had signs of immunodeficiency or failure to thrive at the time of death or had unknown cause of death.  They used this information to determine who may have had SCID.  Based on this chart review the estimated prevalence of SCID is 52/100,000 live Navajo births.

Information from expert interviews
Dr. Rebecca Buckley stated that in one year in North Carolina (where the birth rate is approximately 120,000/annually) there were 3 cases of SCID, for an incidence of 1/40,000.  However, multi-year data confirming this incidence are not available.

Drs. Buckley and Notarangelo provided the distribution of genotypes among the SCID patients for which they have personally cared.  Additionally, Drs Pai, Bonilla, and Notarangelo provided subtype distribution information from both European and United States immunodeficiency collaboratives.  These data corroborated the data from the published literature portion of this evidence review. 

C. What methods exist to screen newborns for SCID?  How accurate are those methods?  What are their sensitivity and specificity?  What methods exist to diagnose individuals with positive screens?

 


Table 6- Quality Assessment of abstracted literature pertaining to Screening Test Characteristics

Overall sensitivity and specificity of screening & false-positive rate

3

Data obtained from screening programs in U.S. population or similar.

0

Data from systematic studies other than from whole population screening.

3

Estimated from the known biochemistry of the condition.

0

 

 

Repeat specimen rate

0

Data obtained from screening programs in U.S. population or similar.

0

Data from systematic studies other than whole population screening.

0

Estimated from the known biochemistry of the condition.

0

 

 

Second-tier testing

1

Data obtained from screening programs in US population or similar.

0

Data from systematic studies other than whole population screening.

1

Estimated from the known biochemistry of the condition.

0

            Adapted from Pandor et al. 2004, Pollitt et al. 1997

At least three different methods of screening for SCID have been proposed including 1) lymphocyte counts of whole blood, 2) quantitative polymerase chain reaction (qPCR), and 3) enzyme linked immunosorbent assay (ELISA) of dried blood spots.  This section will review the evidence regarding screening.  None of the proposed screening methods distinguish the various SCID genotypes and phenotypes.

Table 7- Evaluated literature pertaining to SCID screening studies completed (alphabetical by author)

Study
(Author, Date)

Population

Screening method

Sens, spec, PPV, NPV

Quality of Evidence

Chan, Puck 2005

23 children with SCID
2 children with non-SCID immunodeficiencies

242 anonymized newborn screening cards

DNA amplification of TREC from dried blood spot

Among the children known to have SCID, none had detectable levels of TREC and all had detectable ß-actin.  The 2 children with non-SCID immunodeficiency had detectable TREC.

*False positive rate: 1.5% from routine nurseries;
5% from special-care nurseries.
^Sensitivity: 84%-100%
^Specificity: 97-97.1%

Overall sensitivity and specificity of screening & false-positive rate

 

Hague et al. 1994

135 total children
45 children with SCID
90 children without SCID

matched by age and presenting symptoms with SCID to either asymptomatic or age and presenting symptoms

First available lymphocyte count with cut-off of 2.8x109/l as indicating SCID

Children with SCID had significantly lower levels of lymphocytes.  Unlike the 5 control children with low lymphocyte count, low lymphocyte count persisted in children with SCID

*False-positive rate:  8%
^Sensitivity: 86.3%, and ^Specificity: 94.4%

Overall sensitivity and specificity of screening & false-positive rate

Hennewig et al. 2007

36 children with rotavirus gastroenteritis;
18 with SCID
18 without SCID

Lymphocyte study

SCID children were more likely to have:
low white blood cell count:10/18 vs. 0/18,
eosinophilia: 12/18 vs. 0/18 relative lymphopenia: 17/18 vs. 10/18
absolute lymphopenia:16/18 vs. 4/18

^Sensitivity: 55.6% to 94.4% ^Specificity: 44.4% to 100%. 

Second-tier testing

McGhee et al. 2005

13 children with SCID
183 anonymized dried blood spots, presumed to be from children without SCID

dried blood spot study 

Evaluated a 2-tiered screening approach in which IL-7 is first measured and only those with elevated IL-7 would have TREC measured, although for this study researchers evaluated each test separately.

*Combined specificity of 100% (confidence interval, 97-100%) *Combined sensitivity of at least 85%

Overall sensitivity and specificity of screening & false-positive rate

*Calculation stated in article
^Our calculation using data provided in article

Systematic literature review
Lymphocyte studies: A case-control study (Hague et al. 1994) matched children who were both pre- and post-symptomatically diagnosed with SCID to either asymptomatic children (presenting for surgery or bilirubin screening) or children matched by age and presenting symptoms, in order to compare the earliest available lymphocyte counts. Children with SCID had significantly lower levels of lymphocytes.  Also, unlike in the 5 control children with low lymphocyte counts, in SCID children the low lymphocyte count persisted. The authors calculated a false-positive rate of 8%, positive predictive value of 86% and negative predictive value of 100% among symptomatic children, and a positive and negative predictive value of 93% for all children. As part of the evidence review, we used all data contained within this article to calculate the sensitivity, 86.3%, and specificity, 94.4%, of “first lymphocyte count” using a cut-off of 2.8x109/l as indicating SCID. 
A cohort study (Hennewig et al. 2007) that included 36 children (18 with and 18 without SCID) with rotavirus gastroenteritis, also investigated white blood cell count as a screening tool for SCID.  The researchers found that most of the children with SCID were younger than controls when rotavirus was diagnosed.  They also were more likely to have a low white blood cell count (10/18 vs. 0/18), eosinophilia (12/18 vs. 0/18) and relative lymphopenia (17/18 vs. 10/18) and absolute lymphopenia (16/18 vs. 4/18).  From these numbers we calculated sensitivities from 55.6% to 94.4% and specificities from 44.4% to 100%. 

DBS studies: Two studies published in 2005 evaluated screening methods that utilize dried blood spots. The first (McGhee et al. 2005) evaluated a 2-tiered screening approach in which IL-7 is first measured (Table 8A) and only those with elevated IL-7 would have TREC measured (Table 8B). For the study the researchers evaluated each test separately, rather than using a tiered approach. The investigators tested samples from 13 children with SCID (either dried blood spots [3] collected in immunology clinic or stored serum [10]) and 183 anonymized dried blood spots, presumed to be from children without SCID. They evaluated the levels of T-cell receptor excision circles (TREC) in both groups. However, TREC analysis cannot be done on serum, limiting that portion of the analysis to dried blood spot specimens.

Table 8A- Tier 1: Elevated IL-7 from McGhee et al. 2005

Yes

No

Total

SCID

11

2

13

Control

4

110

114

Total

15

112

127

Authors calculated a specificity of 96.1% for IL-7 and a sensitivity of 85% (confidence interval, 55-98%).

Table 8B- Tier 2: Detectable TREC from McGhee et al. 2005

Yes

No

Total

SCID

0

3

3

Control

169

14

183

Total

169

17

186

Authors calculated a specificity of 92.3% for TREC and a sensitivity that “approaches 100%.”

They evaluated the sensitivity and specificity of the proposed 2-tiered approach calculating a combined specificity of 100% (confidence interval, 97-100%) and combined sensitivity of at least 85%.
Among their sample of SCID patients was one child with Omenn syndrome, which is known to have detectable T-cells, and another patient with T-B- SCID of unknown cause that could theoretically have affected the IL-7 system.  This high proportion of “unusual types” of SCID led the authors to suggest that the actual sensitivity of IL-7 may be higher than shown in this study. 

In the second study (Chan, Puck 2005) investigating screening for SCID using dried blood spots, the sample included 23 children with SCID, 2 children with non-SCID immunodeficiencies, 242 anonymized newborn screening cards.
The researchers used DNA amplification as the screening method in which ß-actin served as a control, indicating that DNA could be amplified from a given specimen, i.e., that the specimen was satisfactory for analysis; and TREC amplification served as the screening test. 
Among the children known to have SCID, none had detectable levels of TREC and all had detectable ß-actin.  The 2 children with non-SCID immunodeficiency had detectable TREC. 
The researchers assumed that the anonymized newborn screening cards came from children without SCID.  In this group there were 7 (2 from routine nursery, 5 from special-care nursery) cards from which TREC could not be detected.  Cards with detectable ß-actin but not TREC were presumed to be false-positive.  They calculated a false-positive rate of 1.5% among children discharged from routine nurseries and 5% among children discharged from special-care nurseries.
We combined the results from this study, except the cards from which ß-actin could not be amplified, and calculated a sensitivity of 84% and specificity of 97.1% for undetectable TREC and sensitivity of 100% and specificity of 97% for TREC <30.

Additional data from expert interviews regarding screening:
Wisconsin implemented universal newborn screening for SCID on January 1, 2008.  Their protocol involves TREC quantification.  For samples with TREC <25/µl they repeat the testing in duplicate, two new punches from the same newborn screening sample collection card, and also assess for ß-actin.  Approximately 1.5% of samples require a second test, and approximately 0.2% requires a second newborn screen or confirmatory testing.  The table below was provided by Dr. Mei Baker as a summary of their screening reports.

 

Table 9- Summary of screening reports for Wisconsin’s universal newborn screening for SCID 

Reporting Type

Situation

Action Plan

Normal

1. TRECs = 25 / ul in the first tier test

None

blank

2.  TRECs = 25 / ul in one or both duplicates of the second tier test

None

Abnormal

TRECs < 25 / ul  and ACTB > 10,000 / ul in full term babies

1.Phone Primary Care Provider and clinical consultant
2. Recommend confirmatory testing

blank

TRECs < 25 / ul  and ACTB > 10,000 / ul in premature infants

1. Phone Primary Care Provider
2. Tracking second NBS*

Inconclusive

TRECs < 25 / ul  and ACTB < 10,000 / ul in full term babies

Recommend repeating NBS

blank

TRECs < 25 / ul  and ACTB < 10,000 / ul in premature infants

Tracking second NBS*

* The second and third NBS in premature infants is a standard practice in the Wisconsin newborn screening program, and no additional sample is requested for the SCID screening test.

As of August 31, 2008, Wisconsin had screened just over 47,250 babies, of which 76 babies had inconclusive screens and 20 had abnormal screens (Table 10 and Table 11).  To date, they have not identified any children with SCID.

Table 10- SCID Screening Results (Courtesy of Dr. Mei Baker, presented at the Newborn Screening Symposium, November, 2008)

  • Number Screened:

47,250 (01/01/2008-08/31/2008)

  • Abnormal Results:

20

    • Premature (<37 weeks)

11 (0.023%)

    • Full term

9 (0.019%)

  • Inconclusive Results

76

    • Premature (<37 weeks)

57 (0.121%)

    • Full term

19 (0.040%)


Table 11- SCID Screening Confirmation Results (Courtesy of Dr. Mei Baker, presented at the Newborn Screening Symposium, November, 2008)


Abnormal Results:

Inconclusive Results:

1 DiGeorge Syndrome
1 Downs Syndrome with sepsis at birth
1 Idiopathic T-cell lymphopenia
1 Leukocyte migration defect
4 normal Flow Cytometry results
9 normal results on repeated newborn screening
2 pending cases
1 expired case

1 DiGeorge Syndrome
59 normal results on repeated newborn screening
2 pending cases
14 expired cases

 

Two other investigators have definitive plans for newborn screening trials.  Dr. Anne Comeau and colleagues at the New England Newborn Screening Laboratory will soon begin a trial of universal newborn screening for SCID in Massachusetts, with a small pilot program in Texas and potential extension to other New England states. Similarly, Dr. Jennifer Puck will soon begin a screening trial at 2 hospitals on Navajo reservations in New Mexico and Arizona, a site chosen because of the high frequency of SCID in the Navajo population. 

Additionally, correspondence with topic experts indicated other screening techniques currently under consideration or evaluation. Drs. Ken Pass indicated that work is underway to develop a T-cell immunoassay that could target key T-cell antigens and serve as a quantitative assay. However, this technique is still under development and has not yet, to our knowledge, been validated. Dr. Anne Comeau indicated that her laboratory has developed a multiplex assay that ensures integral quality assurance; TRECs and RNaseP from the same aliquot of blood are measured in the same reaction.  Furthermore, the assay is amenable to measuring other markers such as key T-cell antigens and development of multi-analyte profiles for SCID.  Proof of principle for the multianlayte assay has been demonstrated; specific non-DNA markers are still in development.

Feasibility/acceptability of screening
We found no articles for inclusion in this review that evaluated the feasibility or acceptability of screening for SCID.

Diagnostic Testing
Review of the literature found no evidence that describes any specific diagnostic testing protocol for SCID.  We suspect this reflects the time-frame used in the literature search and that diagnostic testing protocols were established prior to 1988.

Articles that make reference to diagnostic testing and the experts with whom we spoke all utilize flow cytometry, which allows for the differentiation and quantification of the various sub-types of white blood cells.  As mentioned in conversation with Dr. Puck, the presence of maternal T-cells can confound standard flow cytometry results.  However, further testing for T-cell response to mitogens allows for differentiation between maternal, which do not respond to mitogens in vitro, and child cells.

Additionally, several researchers (Vogt, Puck, Buckley, Notarangelo, Pai and Bonilla) commented on gene sequencing.  The current expert opinion seems to be that knowing the specific genotype, with the exception of ADA-deficiency, has little clinical significance or implications for treatment.  However, the possibility exists that better knowledge of how children with different genotypes respond to treatment may help tailor future treatment.

    • What benefit does treatment, particularly pre-symptomatic, confer?

Numerous studies document that treatment, mainly bone marrow transplant, substantially improves survival. We describe below the evidence regarding the key controversies surrounding forms of treatment and their relative efficacy. We also provide some focus on the evidence regarding early versus later treatment effects. 

Table 12- Quality Assessment of abstracted literature pertaining to Effectiveness of treatment

Effectiveness of treatment

47

I.  Well-designed RCTs.

0

II-1.  Well-designed controlled trials with pseudorandomization or no randomization.

0

II-2.  Well-designed cohort studies:

8

A.  prospective with concurrent controls

0

B.  prospective with historical control

1

C. retrospective with concurrent controls.

7

II-3.  Well-designed case-control (retrospective) studies.

0

III. Large differences from comparisons between times and/or places with and without intervention

4

IV. Opinions of respected authorities based on clinical experience, descriptive studies and reports of expert committees. 

35

Adapted from Pandor et al. 2004, Pollitt et al. 1997

Over the last twenty years, three modes of treatment for SCID have been investigated: allogeneic hematopoietic stem cell transplant (HSCT), the most common subtype being bone marrow transplant (BMT), enzyme replacement therapy (ERT), and gene therapy. While HSCT is the most common treatment for SCID patients, ERT may be used for some patients with ADA-deficient SCID. Lastly, small trials of gene therapy for SCID have been conducted. This section describes the evidence for each method of treatment.

Hematopoietic stem cell transplant-Systematic literature review
HSCT has been utilized as a treatment for SCID since its initial patient application in 1968 (Buckley et al. 1999).  Since then, many researchers have investigated the efficacy, outcomes for different methods, and the long-term outcomes associated with HSCT.  This evidence review focuses on answering the questions pertaining to the benefit of treatment: overall efficacy, efficacy early in life, efficacy for different genotypes and phenotypes of SCID, efficacy based on transplant method, and long-term follow-up including survival and immune reconstitution.

HSCT in the neonatal period and/or infancy

We found 2 studies that specifically addressed HSCT in the neonatal period.  The first was a cohort study (Myers et al. 2002) and the other was a case-series (Kane et al. 2001). In addition, Buckley (Buckley et al. 1999) evaluated a group of patients receiving transplants within the first 3.5 months of life.

Table 13- Abstracted literature pertaining to HSCT in the neonatal period and/or infancy (alphabetical by author)

Study
(Author, Date)

Population

Key Findings

Quality of Evidence

Buckley et al. 1999*

Case series

22 infants transplanted prior to 3.5 months of life
67 infants transplanted later than 3.5 months of life

  • 21/22 (95%) infants alive at follow-up 51/67 (76%) who received transplants at 3.5 months or older survived to follow-up.
  • Median follow-up time was 5.6 years (range 3 months -16.5 years).

IV

Kane et al. 2001

case-series

13 children transplanted between 7 and 68 days-old

  • All patients alive and well 0.5-11.5 years post-transplant (median 3 years).
  • Concluded neonatal transplant has better outcomes than in-utero transplant, but provided no specific data regarding in-utero transplant.
  • 2 children with chronic GvHD chronic.
  • 3 children required more than one transplant.
  • All children achieved: neutrophil engraftment and normal levels of IgA.
  • 7 have normal IgG.
  • 12 have normal IgM.
  • 10/12 maintained normal development, of the other two: 1/12 has developmental problems and 1/12 has motor delay.  

 

 

IV

Myers et al. 2002*

Cohort study

21 children transplanted prior to 28 days of life (early treatment)
96 children transplanted at a median age of 190 days (range 45-516) (late treatment)

  • 20/21 (95%) early treatment children survived.
  • One ADA-deficient patient died of CMV despite successful engraftment.
  • 71/96 (74%) late treatment children survived.
  • Mean time to significant T-cell function in all early treatment was 33 days and to normal T-cell function was 103 days.
  • Mean TREC value peaked earlier post-transplant for early treatment recipients but the 2 groups were indistinguishable by 5 years. 
  • Early transplantation did not have an affect on B-cell function. 
  • 2 early treatment children had HLA-identical donors. 

II-2 C

* Potential patient overlap of Myers et al. 2002, Buckley et al. 1999

Information from expert interviews
Dr. Buckley shared SCID treatment data with our group. They have transplanted 161 SCID infants over the past 26 years, of which 16 had an HLA-identical (genotypically HLA identical) donor. The remaining 145 patients received half-matched (HLA haplo-identical) transplants from their parents. She has documented an overall 26year survival rate of 125/161 (78%). However, her data shows there is a survival difference when the population is divided by age at transplantation. The survival rate in those transplanted before 3.5 months of age is 96%, whereas the survival rate for those transplanted after later is only 71%. All transplants performed and included in this data were done without pre-transplant chemotherapeutic conditioning or post-transplantation GVHD prophylactic immunosuppressive drugs. The Kaplan-Meier graphs from Dr. Buckley (with permission) for these data are below (Graphs 1A and 1B).


Graph 1A- Kaplan-Meier Plot of 48 Children with SCID Transplanted at Duke University Medical Center in the First 3.5 Months of Life

Graph 1A

Graph 1B- Kaplan-Meier Plot of 113 Children with SCID Transplanted at Duke University Medical Center after the First 3.5 Months of Life

Graph 1B
Overview of efficacy—large case-series of HSCT
We found 3 case series studies that specifically addressed the efficacy of HSCT over time.  In one series, 31 patients undergoing BMT for SCID were evaluated between 1968 and 1992 (van Leeuwen et al. 1994). In a similar large case-series (Stephan et al. 1993) 117 patients were followed from 1970 to 1992.  A single-center study (Buckley et al. 1999) followed 89 children between 1982 and 1998.

Table 14- Abstracted literature pertaining to efficacy in large case-series studies of HSCT (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of Evidence

Buckley et al. 1999

Case series

89 children treated with HSCT for SCID between 1982 and 1998

  • 72 (81%) survived to follow-up (median follow-up time was of 5.6 years).
  • HLA-identical transplant from a related donor:  12/12 (100%) survived.
  • T-cell depleted haplo-identical:  60/77 (78%) survived.
  • Survival not statistically related to genotype.
  • Survival varied by race (white more likely to survive) and gender (all girls survived).
  • 36 children developed GVHD.
  • Most cases of GVHD required no treatment. 

 

IV

Stephan et al. 1993*

 

Case series

117  patients treated for SCID between 1970 and 1992; 85 of these children were treated with bone marrow transplant

  • HLA-identical transplant from a related donor 21/25 (84% survived).
  • Pheno-identical transplant (HLA genotypically haplo-identical) from related donor 2/5 (40% survived).
  • HLA haplo-identical transplant without T-cell depletion 0/5 (0% survived).
  • T-cell depleted haplo-identical transplant 28/50 (56% survived).
  • 22 children did not receive any type of transplant and died.
  • 10 received fetal liver transplant, 9 died post transplant.

 

IV

van Leeuwen et al. 1994 *

Case series

31 patients between the ages of 1-94 months undergoing BMT for the treatment of SCID

  • HLA-identical related: 6/10 (60% survived).
  • HLA haplo-identical related: 9/19 (47% survived).
  • HLA-matched unrelated: 0/2 (0% survived).

 

IV

* Potential patient overlap with Stephan et al. 1993 and van Leeuwen et al. 1994

HSCT efficacy in different genotypes and phenotypes of SCID
Several studies examined HSCT for patients with specific pheno- or genotypes, including: B+ SCID vs. B- SCID, SCIDA, reticular dysgenesis, ADA-deficiency, specific Jak3 mutations, and Omenn syndrome. Table 15 summarizes the results of these studies. 

Table 15-Abstracted literature pertaining to HSCT efficacy in different SCID subgroups (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of evidence

1) Albuquerque & Gaspar, 2004

Cohort study
2)
Honig et al. 2007

Case series

 

3)
Rogers et al. 2001

Cohort study

ADA deficiency

1)

  • Compared 12 children with ADA-deficiency to 16 children with other immunodeficiencies, all treated with HSCT.
  • 7/12 ADA-deficient children had bilateral deafness, compared to 1/16 in the control group. 

2)

  • 15 children specifically examined for neurologic outcomes after BMT.
  • 12/15 survived.
  • 6/12 have significant neurocognitive deficits (learning disabilities, gait problems, hearing deficit, and/or hyperactivity). 

3)

  • Compared 11 children treated with HSCT for ADA-deficiency to matched controls treated with HSCT for other forms of SCID.
  • Significant behavioral differences between the 2 groups with ADA-deficient children having more hyperactivity and higher scores for dysfunction in social, emotional and behavioral domains, particularly for older children.
  • ADA-deficient children had a higher rate of deafness. 

1)
II-B

 

 

2)
IV

 

 

3)
II-2 C

Bertrand et al. 1999

Cohort study

B+ SCID vs. B- SCID

  • Disease-free survival was better for B+ SCID (60% vs. 35% at mean follow up of 57 and 52 months, respectively).
  • Reduced survival among children with B- SCID was associated with higher incidences of death from infection and chronic GVHD, as well as lower rates of marrow engraftment. 
  • Among B- SCID cases, the method of T-cell depletion significantly impacted the cure rate. 
  • When analyzed as pre- vs. post-1991, both groups had better survival post-1991, but B+ SCID still did better.

II-2 C

Bertrand et al. 2002

Case series

Reticular Dysgenesis

 

  • 3/10 patients with reticular dysgenesis treated with HLA-non-identical HSCT survived.
  • Most deaths related to failure of engraftment.
  • Rates of engraftment appeared to be better among children who received conditioning chemotherapy prior to transplant.

IV

Gomez et al. 1995

Case series

Omenn syndrome

  • 9 children with Omenn syndrome receiving BMT after nutritional supplementation and immunosuppression. 
  • Engraftment occurred in 4/5 HLA-identical transplants and 3/4 non-identical. 
  • 3 children developed GVHD and one died of CMV 50 days post-transplant.
  • Clinical manifestations of Omenn syndrome disappeared within days after BMT.
  • At follow-up, all survivors except one with chronic GVHD have normal growth rates.

IV

O'Marcaigh et al. 2001

Case series

SCIDA

  • No deaths in first 100 days after transplant for 16 SCIDA patients.
  • 15/16 achieved engraftment.
  • 11/16 developed normal T cell function.

IV

Roberts et al. 2004

Case series

Jak3 mutations

  • 9 different Jak3 mutations in 10 different SCID patients. 
  • 9/10 were alive and without recurrent infections 4-18 years after transplant.

IV

HSCT variation in transplant methods

Multiple studies compare various transplant methods and their outcomes.  The main points of discussion surround the degree of matching between the donor and recipient, the extent of myeloablation prior to transplant and the method for T-cell depletion of the donor stem cells.  In addition, investigators have considered the source of stem cells and the use of “booster transplants”.

The extent of matching between the donor and recipient is only partially under the control of physicians, as many patients lack an available matched related donor. This results in recipients requiring either a haplo-identical or mismatched transplant.  Several studies have looked at the various types of transplants and their efficacy.

Table 16- Abstracted literature pertaining to HLA matching and efficacy of HSCT (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of evidence

Dalal et al. 2000

Case series

16 children, 9 with SCID, who lacked histocompatible siblings or closely matched related donors between  1989 and1997    

  • 12 patients survived.
  • Neutrophil engraftment was achieved in all patients at a mean of 15.4 days.
  • Serum IgM and IgA normalized in all patients within a mean of 3.5 and 6 months.
  • 2/9 (22%) SCID patients developed GVHD and died.

IV

Dal-Cortivo et al. 2004

Case series

25 children in which there was no potential to exert NK-cell alloreactivity compared to 13 in which there was such potential (defined by donor expression of inhibitory killer immunoglobulin-like receptor (KIR)), all receiving haploidentical transplants

  • No difference in the rates of engraftment (64% vs. 61.5%).
  • Trend towards lower incidence of grade II-IV acute GVHD in the patients with potential NK-cell alloreactivity (37.5% vs. 50%, p=0.68). 
  • One year survival was not significantly different between the two groups (52% vs. 61.5%).

IV

Giri et al. 1994

Case series

11 children receiving HLA-non-identical BMT between 1985 and 1992

  • 9 patients engrafted.
  • 5 (46%) patients survived 6-78 months post-BMT.
  • 4/9 developed GVHD.

IV

Grunebaum et al. 2006

Cohort study

Compared recipient outcomes between:
- related HLA-identical donors (RID, n=13),
- HLA-matched unrelated donors (MUD, n= 41)
- HLA-mismatched related donors (MMRD, n=40). 

  • Median time to transplant was 1 month for RID, 2 months for MMRD and 4 months for MUD.
  • Highest survival was RID (92.3%), followed by MUD (80.5%) and MMRD (52.5%). 
  • 0% of RID, 7.3% of MUD and 30% of MMRD had graft failure.
  • No statistical difference between MUD and MMRD lymphocyte subsets and lymphocyte function.
  • Patient sex and presence of B cells has no impact on survival. 
  • Respiratory complications occurred in 7.3% of MUD and 35% of MMRD; no respiratory complications in RID children.
  • GVHD most common following MUD BMT (73.1%) or MMRD BMT (45%). 

II-2 C

Smogorzewska et al. 2000

Cohort study

11 children treated with histocompatible transplants from a sibling and
37 children treated with T-cell depleted haploidentical parental bone marrow between 1984 and 1997

  • 100% survival in histocompatible group.
  • 46% survival in haploidentical group.
  • Mean age at transplant of children who survived: 7.5 months.
  • Mean age at transplant of children who died: 11.4 months.
  • All surviving children recovered T-cell function.
  • Recovery slower for children receiving haploidentical transplants.

II-2 C

Wijnaendts et al. 1989

Case series

33 children who received transplants between 1972 and 1987 and survived a minimum of 6 months:18 HLA-identical and 15 HLA-non-identical

  • Development of immune function occurred faster in patients with HLA-identical transplants. 
  • Development of T- and B-cell function occurred faster when chemotherapy preceded BMT.
  • Poor B-cell function was observed more frequently when chemotherapy was not used.

IV

 

Table 17- Abstracted literature pertaining to the role of myeloablation prior to transplant (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of evidence

Amrolia et al. 2000

Case series

Children ineligible for conventional myeloablation due to comorbidities; 8 patients, including 5 with SCID

  • All treated with non-myeloablative chemotherapy prior to transplant.
  • 7/8 children survived.
  • At median follow-up of 12 months: all 7 survivors have good recovery of T-cell numbers, 4 patients have normal IgM levels, and 2 patients have normal IgA levels.

 

IV

Rao et al. 2005

Cohort study

Compared 33 children receiving reduced intensity pre-transplant chemotherapy and 19 receiving myeloablation. 

  • All children in both groups had primary engraftment.
  • Reduced intensity group: 32/33 were alive at one month and 31/33 alive at one year
  • Myeloablative group: 14/19 were alive at one month and 11/19 alive at one year. 
  • At one year: all in the myeloablative group had normal B-cell function, 5 of reduced intensity group still required IVIG. 
  • GVHD incidence not different between groups but limited chronic was more common in myeloablative group.
  • Quality of life, as measured by Lansky score, was similar between the groups (97 vs. 94).

III

Veys, Rao & Amrolia 2005

Case series

81 children with congenital immunodeficiencies, 20 with SCID

  • All subjects underwent HSCT with reduced-intensity conditioning.
  • Survival rate of 84% (68/81).
  • Authors concluded that HSCT with reduced intensity conditioning was well tolerated.

IV

      Other variation in treatment methods

Three studies specifically investigated stem cell sources other than bone marrow.  A small study (Knutsen, Wall 2000) which included only 8 children with SCID evaluated transplants using umbilical cord blood derived stem cells and found neutrophil recovery time was the same as that reported in BMT, while platelet recovery was delayed compared to BMT.  A study from Turkey (Arpaci et al. 2008) used peripheral stem cells mobilized by treating donors with G-CSF.  21 patients, of whom 16 had SCID, received a total of 28 haploidentical HSCT.  Median age at transplant was 12 months.  At last follow-up 8/21 children were alive with the authors believing that delay in transplantation accounted for the poor outcomes compared to other studies.  Finally, stem cells from fetal liver (Touraine et al. 2007) were used for transplants in 17 infants; in the 3 patients for whom information was available, they found normal antibody responses several years after transplant.
One study (Dror et al. 1993) reported on 24 patients who underwent a total of 36  using lectin-treated t-cell depleted haplocompatible BMT.  Of these 24 children durable T-cell engraftment was achieved in 19, although 6/19 required more than one transplant to achieve engraftment.  Pre-transplant conditioning positively affected engraftment, while cell dose, patient’s age, and donor and patient sex had no observable effect on engraftment rates

      For patients that achieve engraftment but not adequate immune function, bone marrow boosts (Kline, Stiehm & Cowan 1996), in which additional stem cells from the same donor were given without pre-treatment, led to increased absolute lymphocyte counts in 5/9, increased mean CD3, CD4 and CD8 counts and improved mean T-cell function. Mentioned prior, the long-term follow up case-series (Buckley et al. 1999) also included a total of 20/89 receiving “booster” transplants to overcome poor T/B cell function or poor engraftment. Of those who received a booster transplant, immune function improved in all but 3. 

HSCT long-term follow up, survival and immune reconstitution

Table 18- Abstracted literature pertaining to long-term follow-up and survival after HSCT (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of evidence

Antoine et al. 2003 *

Cohort study

475 patients (total of 566 transplants) from 37 European centers between 1968 and 1999

  • Three-year survival with sustained engraftment was 77% for HLA-identical and 54% for HLA-non-identical transplants. 
  • Survival has improved over time for both HLA-identical and HLA-non-identical transplant recipients. 
  • B- SCID (36% 3-yr survival; CI 26-45%) had a poorer prognosis than B+ SCID (64% 3-yr survival; CI  57-72%).
  • Myeloablation prior to HLA-nonidentical transplant trended towards improving survival among B- children but not B+. 
  • ADA-deficiency (n=51) 3-yr-survival was 81% for matched and 29% for unmatched transplants.   
  • Reticular dysgenesis survival (n=12) was 75% for matched and 29% for unmatched.

II-2 C

Cavazzana-Calvo et al. 2007

Case series

31 children 10-27 years post-transplant: focus on factors associated with long-term T-cell reconstitution. 

  • Myeloablation patients more likely to have evidence of donor-derived granulocytes and persistent naïve T-cells, as measured by TREC.
  • Average follow-up time in the TREC+ group was 13 years and in the TREC- was 16 years. 
  • At follow-up, 60% of TREC+ and 45% of TREC- had no clinical manifestations.

IV

Fischer et al. 1990 *

Case series

475 patients (total of 566 transplants) gathered between 1968 and 1999 from 37 European centers

 

  • Survival significantly better for HLA-identical (76% survival) than HLA-non-identical transplants (56% survival). 
  • SCID phenotype was not associated with difference in survival.
  • Lung infection before HSCT and absence of a protective environment significantly affected outcome (multivariate analysis).
  • A total of 27% had acute GVHD of grade II or higher and 25% developed chronic GVHD.

IV

Friedrich, Honig & Muller 2007

Cohort study

 7 children with SCID receiving HLA-identical transplant, 25 with HLA-haploidentical transplant, all at least 10 years out from transplant

  • Most patients had normal and stable T-cell numbers and functions.
  • 3 patients’ T-cell numbers were decreased.
  • 4 patients’ PHA responses were decreased (all in HLA-haploidentical group and no chemotherapy). 
  • HLA-haploidentical with no conditioning had lower levels of naïve CD4+ cells and impaired B cell functioning.

IV

Gennery et al. 2001

Case series

19 children treated from 1987-1998 using CAMPATH-1MT depleted BMT for SCID

  • 19/30 children survived longer than 1 year post-BMT
  • Most deaths attributed to pre-existing infection.
  • 17 had normal immune function following transplantation.

IV

Haddad et al. 1998

Case series

193 patients from18 European centers between 1982 and 1993: focus on immune reconstitution

  • 116 alive with evidence of engraftment 5 months after BMT. 24 later died (20%).
  • T-cell function improved during the 2 years after BMT and continued to be better than B-cell function. 
  • Poor outcomes associated with: absence of T-cell reconstitution, presence chronic GVHD 6 months after transplant, B- SCID (multivariate analysis).
  • At last follow up (median, 6 years after transplant), 93% had normal T-cell function and 68% had normal B-cell function. 

 

IV

Slatter et al. 2008

Cohort study

36 children treated with BMT with depletion of T-cells from a non-identical donor 

  • No significant survival difference between children receiving transplants depleted using anti-CD52 or anti-CD34 antibodies.
  • 5 patients in the anti-CD52 group and 2 in anti-CD34 group had GvHD.

II-2 C

*A subset of patients in Fischer et al. 1990 are also included in Antoine et al. 2003

Table 19- Abstracted literature pertaining to long-term immune reconstitution after HSCT (alphabetical by author)

Study
(Author, Date)

Population

Key findings

Quality of evidence

Borghans et al. 2006

Case control study

19 people treated with BMT for SCID (5-32 years earlier) compared to 173 healthy controls. 

  • Median number of T-cells was lower in healthy matched controls: 11 SCID patients had normal T-cell counts.
  • 8 SCID patients who had low t-cell counts at last follow-up also had low counts early (1-4 years after BMT).
  • Predictors of low T-cell count included NK+ SCID, B- SCID, earlier transplant.
  • No significant difference in late clinical outcomes found between patients with and without “good” immune reconstitution.

III

Brugnoni et al. 1998

Case series

8 childrens’ immune reconstitution and  response to PHA after BMT from unrelated donors

  • Number of activated T-cells decreased in the months after BMT.
  • Proliferative response to PHA initially decreased and then increased to normal by 8 months post-transplant.

IV

Mazzolari et al. 2007

Case series

58 children treated with HSCT (in-utero to 34 months at time of transplant)

  • 42/58 (72.4%) survived at least 5 years (median follow-up of 132 months). 
  • 85% appropriately produced antibodies to vaccines (tetanus toxoid and hepatitis B).
  • 26/28 immunized with Measles, Mumps and Rubella vaccine had evidence of anti-measles antibodies.
  • Most children had height (77.5%) and weight (82.5%) in the normal range.
  • At last follow-up, 24/40 required no treatment, 6 required IVIG and/or antibiotics, 5 are on thyroid replacement, 3 receive anti-epileptics and 1 is treated for portal hypertension.

IV

Patel et al. 2000

Case series

173 total patients: 83 SCID and 90 normal Mean age of transplant was 0.5 ±0.4 years; controls ranged from <1 to 79 years

  • Non-thymus-dependent cells predominated in the first 100 days after transplant.
  • At 140-180 days after transplant thymus-dependent cells predominated. 
  • After transplant thymus-dependent cells peaked at approximately one year but then diminished over the next 14. 
  • Normal controls showed a decline in thymus-dependent cells from birth to age 79 years.

IV

Vossen et al. 1993

Case series

14 children evaluated 1-23 years after BMT 

  • All children who survived greater than 1 year had some donor T-cell engraftment.
  • Cellular immunity was quantitatively low but mostly functionally normal.

IV

 

Information from expert interviews
Consultation with experts pertaining to treatment of SCID corroborated the data from the published literature portion of this evidence review. Experts concurred that major challenges to treating SCID are due to complications prior to diagnosis and treatment (such as infections), and a lack of a uniform approach for treatment. Drs. Buckley and Notarangelo provided information on development of two SCID consortiums: United States Immunodeficiency Network (USIDNET) Registry sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) and Center for International Blood and Marrow Transplant Research (CIBMTR). While USIDNET is focused on the registry of primary immune deficiencies, reporting of all BMT procedures to the CIBMTR registry has been mandatory since December. Data from these consortiums may, in the future, provide more data on treatment outcomes for patients with SCID.

At this time, Dr. Buckley reports there are fifteen major and 34 minor centers in the U.S. and Canada currently performing stem cell transplantation for SCID. Dr. Notarangelo, Bonilla and Pai stated that an informal survey performed under the auspices of the NIAID/Rare Diseases workshop identified 34 centers in the United States and Canada that currently perform HCT for SCID (unpublished data, NIAID May 12-13, 2008 HCT for Primary Immune Deficiency Diseases workshop).

Enzyme replacement therapy-Evidence Review
Enzyme replacement therapy has been a therapeutic option for patients with ADA-deficiency, a specific type of SCID.  Although multiple case studies examining the use of polyethylene glycol- adenosine deaminase (PEG-ADA) have been published, our examination of the literature found only one study meeting criteria for inclusion in this evidence review.
A long-term follow up study by Chan et al (Chan et al. 2005), evaluated outcomes in 9 children treated with PEG-ADA.  These children were diagnosed with ADA-deficiency at ages ranging from birth to 6.5 years and were followed on intramuscular PEG-ADA for 5-12 years.  Some patients also received other treatments. One child died after 9 yrs of PEG-ADA treatment and a failed bone marrow transplant.  The remaining 8 children attended school without protective precautions although 2 children have disabilities thought to be related to early, severe infections.  Lymphocyte counts (T, B and NK) and function increased significantly following initiation of PEG-ADA and peaked at 1-3 years after treatment initiation. Over time lymphocyte counts and T-cell function, as measured by PHA mitogen stimulation have diminished.

Gene therapy-Evidence Review
Gene therapy using viral vectors has been tried for the treatment of X-linked SCID and ADA-deficiency SCID. Two case-series of patients treated with gene therapy were included in this evidence review. 
In 2002, Hacein-Bey-Abina et al (Hacein-Bey-Abina et al. 2002) published a report of 5 boys with X-linked SCID due to a mutation in the common gammac chain gene who were treated by infusing them with their own bone marrow which had previously been extracted and transduced with a vector containing gammac chain derived from a defective Moloney murine leukemia virus.  4/5 had clinical improvement including resolution of infections, diarrhea and skin lesions.  At follow-up 0.7-2.5 years later those 4 were well with normal growth.  The fifth patient never had reconstitution of T-cells following gene therapy and underwent a bone marrow transplantation 8 months later.  With regards to immunologic function, 3/5 patients had normal T-cell values 3 to 4 months after gene therapy.  At last follow up, T-cells from 4/5 patients exhibited normal proliferative responses to in vitro stimulation with phytohemagglutinin and anti-CD3 antibody.   
Schmidt et al (Schmidt et al. 2005) reported on a series of 10 patients (5 of whom were included in the Hacein-Bey-Abina paper) with a focus on longer-term follow-up after gene therapy.  They found polyclonal T-cell repertoires, in the 9/10 patients who developed normal T-cell counts after treatment, indistinguishable from those of age-matched controls. In long-term follow up, 2/9 developed monoclonal lymphoproliferation 2.5 years post transplant.   Additionally, the number of circulating naïve T-cells was similar to that for age-matched controls.

    • Benefits of treatment in screen positive children

We found no evidence related to the benefits of treatment in children who screen positive for SCID.  The relative outcomes for early treatment versus late treatment of children with SCID provide some evidence of improved outcomes with earlier treatment.

    • Harms of SCID screening

We found no evidence related to harms associated with screening for SCID.

    • Harms of SCID diagnosis

We found no evidence related to harms associated with diagnosing SCID.


    • Harms of SCID treatment

 

We found two papers specifically focused on a harm associated with treatment of SCID. The first (Horn et al. 1999) analyzed the rates of auto-immune hemolytic anemia in children undergoing HSCT for SCID. 8/41 children developed auto-immune hemolytic anemia, 3 died from its complications. This was a higher rate of auto-immune hemolytic anemia than previously reported and higher than the rate, in the same institution, among patients undergoing HSCT for non-SCID diseases. Among several potential predictors for the development of auto-immune hemolytic anemia that were analyzed, only peripheral blood as source of stem cells was found to be related to increased risk of developing auto-immune hemolytic anemia. 
Work by Hacein-Bey-Abina et al (Hacein-Bey-Abina et al. 2008) documented a total of 4 children (of the 9/10 who had successful treatment with gene therapy) who developed leukemia between 30 and 68 months after gene therapy.  All cases were associated with vector insertion near genes associated with cancer development.  3/4 kids were successfully treated with chemotherapy and regained poly-clonal T-cell populations; the 4th child underwent 2 BMTs and ultimately died 60 months after gene therapy and 26 months after leukemia diagnosis.

    • Cost-Effectiveness

 

Table 20- Quality Assessment of abstracted literature pertaining to Economic evidence

Economic evidence

1

I.  Evaluation of important alternative interventions comparing all clinically relevant outcomes against appropriate cost measurement and including a clinically sensible sensitivity analysis.

0

II.  Evaluation of important alternative interventions comparing a limited number of outcomes against appropriate cost measurement, but including a clinically sensible sensitivity analysis.

0

III.  Evaluation of important alternative interventions comparing all clinically relevant outcomes against inappropriate cost measurement, but including a clinically sensible sensitivity analysis.

1

IV.  Evaluation without a clinically sensible sensitivity analysis

0

V.  Expert opinion with no explicit critical appraisal, based on economic theory

0

Adapted from NHS Centre for Reviews and Dissemination Report 4, March 2001

We found one study (McGhee, Stiehm & McCabe 2005) that addressed the cost-effectiveness of SCID screening, as it would apply to the United States population.  Using a deterministic decision-tree model, comparing universal and targeted screening approaches, the authors assessed the thresholds at which screening would be cost-effective from a health care system perspective.  They noted a significant amount of uncertainty in their model parameters due, in part, to the lack of SCID screening studies.  Additionally, utility estimates were based on information from patients receiving HSCT for oncologic processes.  The authors found that, at a threshold of $100,000 per quality adjusted life year, there is 86% likelihood of screening being cost-effective, but details of the Monte Carlo simulation used to arrive at this estimate were not provided in the paper. 

Data from experts regarding cost-effectiveness
None of the experts with whom we had contact provided any specific cost-effectiveness information.  However, several had collected some hospital cost or charge data for patients undergoing HSCT for SCID.   These costs were generally at least two to three times higher than those used by McGhee et al in their base-case analysis, suggesting that the costs used in the paper may not reflect current treatment practice.  However, the base-case analysis costs of testing are consistent with the current testing costs in the screening trials.

VIII.  SUMMARY

Key findings:

Severe Combined Immunodeficiency (SCID) affects at least 1/100,000 newborns within the United States.  However, experts believe that with systematic case-finding the prevalence may be higher (perhaps as high as 1/40,000) due to earlier diagnosis of infants who would otherwise die prior to confirming a diagnosis of SCID.  Although several population-based screening trials are underway (Wisconsin) or planned (Massachusetts, Navajo Reservation in Arizona and New Mexico), to date no population-based screening trial has been completed.

Without curative treatment, newborns with SCID develop severe, often opportunistic, infections which lead to early death.  Studies indicate that treatment, most commonly with hematopoietic stem cell transplant, is effective in decreasing both the morbidity and mortality associated with SCID.  Additionally, despite the lack of population-based screening, there is some evidence that earlier treatment may lead to better outcomes.

Regarding the key questions:

  • Do current screening tests effectively and efficiently identify cases of SCID?

Various screening methods ranging from targeted use of lymphocyte counts in hospitalized children to population based newborn screening using IL-7, TREC or a combination have been studied.  Both IL-7 and TREC have been investigated using anonymized dried-blood spots.  These studies allow for estimations of sensitivity and specificity, but due to the anonymous nature there is no way to prove which test results are true.

The state of Wisconsin began a trial of population-based screening for SCID, using low TREC as the marker of SCID, in January 2008.  As of August 31 they had screened 47,250 newborns of which 76 (0.161%) had initially inconclusive results requiring a second newborn screening sample and 20 (0.042%) had abnormal results requiring either a second newborn screening sample or diagnostic testing.  The rates for both inconclusive and abnormal results are higher in premature than in full-term infants.  As of August 31, the investigators have not detected any cases of SCID.

  • Does pre-symptomatic or early symptomatic intervention in newborns or infants with SCID improve health outcomes?

The most common, and well-studied, treatment modality for SCID is hematopoietic stem cell transplant which appears to be effective in significantly decreasing the morbidity and mortality associated with SCID.  Within HSCT there are numerous variables which may contribute to the effectiveness.  The most studied of these are the degree of haplotype matching between donor and recipient and the type of pre-conditioning the recipient receives.  There is no clear evidence as to the best method for HSCT.  However, despite the lack of randomized controlled trials of SCID treatment, the evidence from large case-series indicates that HSCT is effective in treating SCID. 
Furthermore, there is limited evidence, primarily from the early detection of siblings of known SCID cases, that earlier treatment may be more effective. Specifically, the existing evidence indicates that undergoing HSCT prior to the onset of lung infection is more likely to be successful.

In addition to HSCT, there is limited evidence that for children with ADA-deficiency SCID enzyme replacement may be effective in reconstituting their immune function.

  • What is the cost-effectiveness of newborn screening for SCID?

The cost-effectiveness data reviewed was very limited and may not reflect current costs of treatment.

  • What critical evidence appears lacking that may inform screening recommendations for SCID?

We identified several areas with deficient data:

          • Prevalence of SCID

There is limited evidence regarding the true prevalence of SCID.  A systematic method of case finding is needed in order to accurately determine the prevalence.  Of note, the new consortium of treatment centers (USIDNET) that has recently been established may serve as a method of more systematic case-finding.

          • Accuracy of Screening

Initial pilot screening data from Wisconsin suggests that the false-positive rate will be relatively low.  However, these data are limited at this time.  Data regarding the accuracy of other screening methods, when applied in population-based protocols, are not available.

          • Feasibility of Screening

Wisconsin has been able to implement universal newborn screening for SCID, at least on a pilot basis.  Data are needed regarding the ability of other newborn screening laboratories to offer SCID screening

          • Acceptability of Screening

There are no data available regarding consumer or physician acceptance of newborn screening for SCID.

          • Cost-effectiveness

Cost-effectiveness analyses utilizing measured costs and utilities, as well as applicable sensitivity analyses, are needed.

          • Adequacy of available treatment centers

There is no data addressing variation in treatment success between centers or the number of centers in the United States and their capacity to provide treatment for SCID.  Data from USIDNET and CIBMTR may, in the future, provide evidence on this topic.

 


 

IX. REFERENCES
"Primary immunodeficiency diseases. Report of a WHO Scientific Group", 1995, Clinical and experimental immunology, vol. 99 Suppl 1, pp. 1-24.
Severe Combined Immunodeficiency [Homepage of U.S. National Library of Medicine and the National Institutes of Health], [Online]. Available: [2008, October] .
Albuquerque, W. & Gaspar, H.B. 2004, "Bilateral sensorineural deafness in adenosine deaminase-deficient severe combined immunodeficiency", The Journal of pediatrics, vol. 144, no. 2, pp. 278-280.
Amrolia, P., Gaspar, H.B., Hassan, A., Webb, D., Jones, A., Sturt, N., Mieli-Vergani, G., Pagliuca, A., Mufti, G., Hadzic, N., Davies, G. & Veys, P. 2000, "Nonmyeloablative stem cell transplantation for congenital immunodeficiencies", Blood, vol. 96, no. 4, pp. 1239-1246.
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., European Group for Blood and Marrow Transplantation & European Society for Immunodeficiency 2003, "Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: report of the European experience 1968-99", Lancet, vol. 361, no. 9357, pp. 553-560.
Arpaci, F., Tezcan, I., Kuzhan, O., Yalman, N., Uckan, D., Kurekci, A.E., Ikinciogullari, A., Ozet, A. & Tanyeli, A. 2008, "G-CSF-mobilized haploidentical peripheral blood stem cell transplantation in children with poor prognostic nonmalignant disorders", American Journal of Hematology, vol. 83, no. 2, pp. 133-136.
Bertrand, Y., Landais, P., Friedrich, W., Gerritsen, B., Morgan, G., Fasth, A., Cavazzana-Calvo, M., Porta, F., Cant, A., Espanol, T., Muller, S., Veys, P., Vossen, J., Haddad, E. & Fischer, A. 1999, "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", The Journal of pediatrics, vol. 134, no. 6, pp. 740-748.
Bertrand, Y., Muller, S.M., Casanova, J.L., Morgan, G., Fischer, A. & Friedrich, W. 2002, "Reticular dysgenesis: HLA non-identical bone marrow transplants in a series of 10 patients", Bone marrow transplantation, vol. 29, no. 9, pp. 759-762.
Boeck, A., Buckley, R.H. & Schiff, R.I. 1997, "Gastroesophageal reflux and severe combined immunodeficiency", The Journal of allergy and clinical immunology, vol. 99, no. 3, pp. 420-424.
Borghans, J.A., Bredius, R.G., Hazenberg, M.D., Roelofs, H., Jol-van der Zijde, E.C., Heidt, J., Otto, S.A., Kuijpers, T.W., Fibbe, W.E., Vossen, J.M., Miedema, F. & van Tol, M.J. 2006, "Early determinants of long-term T-cell reconstitution after hematopoietic stem cell transplantation for severe combined immunodeficiency", Blood, vol. 108, no. 2, pp. 763-769.
Boyle, J.M. & Buckley, R.H. 2007, "Population prevalence of diagnosed primary immunodeficiency diseases in the United States", Journal of clinical immunology, vol. 27, no. 5, pp. 497-502.
Brugnoni, D., Airo, P., Malagoli, A., Cattaneo, R., Pennacchio, M. & Porta, F. 1998, "Rapid regeneration of normally functional naive CD4(+) T cells after bone marrow transplantation from unrelated donors for combined immunodeficiency", Blood, vol. 92, no. 9, pp. 3484-3486.
Buckley, R.H., Schiff, R.I., Schiff, S.E., Markert, M.L., Williams, L.W., Harville, T.O., Roberts, J.L. & Puck, J.M. 1997, "Human severe combined immunodeficiency: genetic, phenotypic, and functional diversity in one hundred eight infants", The Journal of pediatrics, vol. 130, no. 3, pp. 378-387.
Buckley, R.H., Schiff, S.E., Schiff, R.I., Markert, L., Williams, L.W., Roberts, J.L., Myers, L.A. & Ward, F.E. 1999, "Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency", The New England journal of medicine, vol. 340, no. 7, pp. 508-516.
Cavazzana-Calvo, M., Carlier, F., Le Deist, F., Morillon, E., Taupin, P., Gautier, D., Radford-Weiss, I., Caillat-Zucman, S., Neven, B., Blanche, S., Cheynier, R., Fischer, A. & Hacein-Bey-Abina, S. 2007, "Long-term T-cell reconstitution after hematopoietic stem-cell transplantation in primary T-cell-immunodeficient patients is associated with myeloid chimerism and possibly the primary disease phenotype", Blood, vol. 109, no. 10, pp. 4575-4581.
Chan, B., Wara, D., Bastian, J., Hershfield, M.S., Bohnsack, J., Azen, C.G., Parkman, R., Weinberg, K. & Kohn, D.B. 2005, "Long-term efficacy of enzyme replacement therapy for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID)", Clinical immunology (Orlando, Fla.), vol. 117, no. 2, pp. 133-143.
Chan, K. & Puck, J.M. 2005, "Development of population-based newborn screening for severe combined immunodeficiency", The Journal of allergy and clinical immunology, vol. 115, no. 2, pp. 391-398.
Dalal, I., Reid, B., Doyle, J., Freedman, M., Calderwood, S., Saunders, F. & Roifman, C.M. 2000, "Matched unrelated bone marrow transplantation for combined immunodeficiency", Bone marrow transplantation, vol. 25, no. 6, pp. 613-621.
Dal-Cortivo, L., Ouachee-Chardin, M., Hirsch, I., Blanche, S., Fischer, A., Cavazzana-Calvo, M. & Caillat-Zucman, S. 2004, "Does haploidentical transplantation in children with primary immunodeficiencies have the potential to exploit donor NK cell alloreactivity?", Bone marrow transplantation, vol. 34, no. 11, pp. 945-947.
Deerojanawong, J., Chang, A.B., Eng, P.A., Robertson, C.F. & Kemp, A.S. 1997, "Pulmonary diseases in children with severe combined immune deficiency and DiGeorge syndrome", Pediatric pulmonology, vol. 24, no. 5, pp. 324-330.
Denianke, K.S., Frieden, I.J., Cowan, M.J., Williams, M.L. & McCalmont, T.H. 2001, "Cutaneous manifestations of maternal engraftment in patients with severe combined immunodeficiency: a clinicopathologic study", Bone marrow transplantation, vol. 28, no. 3, pp. 227-233.
Diaz de Heredia, C., Ortega, J.J., Diaz, M.A., Olive, T., Badell, I., Gonzalez-Vicent, M. & Sanchez de Toledo, J. 2008, "Unrelated cord blood transplantation for severe combined immunodeficiency and other primary immunodeficiencies", Bone marrow transplantation, vol. 41, no. 7, pp. 627-633.
Dror, Y., Gallagher, R., Wara, D.W., Colombe, B.W., Merino, A., Benkerrou, M. & Cowan, M.J. 1993, "Immune reconstitution in severe combined immunodeficiency disease after lectin-treated, T-cell-depleted haplocompatible bone marrow transplantation", Blood, vol. 81, no. 8, pp. 2021-2030.
Fischer, A., Landais, P., Friedrich, W., Morgan, G., Gerritsen, B., Fasth, A., Porta, F., Griscelli, C., Goldman, S.F. & Levinsky, R. 1990, "European experience of bone-marrow transplantation for severe combined immunodeficiency", Lancet, vol. 336, no. 8719, pp. 850-854.
Friedrich, W., Honig, M. & Muller, S.M. 2007, "Long-term follow-up in patients with severe combined immunodeficiency treated by bone marrow transplantation", Immunologic research, vol. 38, no. 1-3, pp. 165-173.
Gennery, A.R., Dickinson, A.M., Brigham, K., Barge, D., Spickett, G.P., Curtis, A., Spencer, V., Jackson, A., Cavanagh, G., Carter, V., Palmer, P., Flood, T.J., Cant, A.J. & Abinun, M. 2001, "CAMPATH-1M T-cell depleted BMT for SCID: long-term follow-up of 19 children treated 1987-98 in a single center", Cytotherapy, vol. 3, no. 3, pp. 221-232.
Giri, N., Vowels, M., Ziegler, J.B., Ford, D. & Lam-Po-Tang, R. 1994, "HLA non-identical T-cell-depleted bone marrow transplantation for primary immunodeficiency diseases", Australian and New Zealand Journal of Medicine, vol. 24, no. 1, pp. 26-30.
Gomez, L., Le Deist, F., Blanche, S., Cavazzana-Calvo, M., Griscelli, C. & Fischer, A. 1995, "Treatment of Omenn syndrome by bone marrow transplantation", The Journal of pediatrics, vol. 127, no. 1, pp. 76-81.
Grunebaum, E., Mazzolari, E., Porta, F., Dallera, D., Atkinson, A., Reid, B., Notarangelo, L.D. & Roifman, C.M. 2006, "Bone marrow transplantation for severe combined immune deficiency", JAMA : the journal of the American Medical Association, vol. 295, no. 5, pp. 508-518.
Hacein-Bey-Abina, S., Garrigue, A., Wang, G.P., Soulier, J., Lim, A., Morillon, E., Clappier, E., Caccavelli, L., Delabesse, E., Beldjord, K., Asnafi, V., MacIntyre, E., Dal Cortivo, L., Radford, I., Brousse, N.,Sigaux, F., Moshous, D., Hauer, J., Borkhardt, A., Belohradsky, B.H., Wintergerst, U., Velez, M.C., Leiva, L., Sorensen, R., Wulffraat, N., Blanche, S., Bushman, F.D., Fischer, A. & Cavazzana-Calvo, M. 2008, "Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1", The Journal of clinical investigation, vol. 118, no. 9, pp. 3132-3142.
Hacein-Bey-Abina, S., Le Deist, F., Carlier, F., Bouneaud, C., Hue, C., De Villartay, J.P., Thrasher, A.J., Wulffraat, N., Sorensen, R., Dupuis-Girod, S., Fischer, A., Davies, E.G., Kuis, W., Leiva, L. & Cavazzana-Calvo, M. 2002, "Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy", The New England journal of medicine, vol. 346, no. 16, pp. 1185-1193.
Haddad, E., Landais, P., Friedrich, W., Gerritsen, B., Cavazzana-Calvo, M., Morgan, G., Bertrand, Y., Fasth, A., Porta, F., Cant, A., Espanol, T., Muller, S., Veys, P., Vossen, J. & Fischer, A. 1998, "Long-term immune reconstitution and outcome after HLA-nonidentical T-cell-depleted bone marrow transplantation for severe combined immunodeficiency: a European retrospective study of 116 patients", Blood, vol. 91, no. 10, pp. 3646-3653.
Hague, R.A., Rassam, S., Morgan, G. & Cant, A.J. 1994, "Early diagnosis of severe combined immunodeficiency syndrome", Archives of Disease in Childhood, vol. 70, no. 4, pp. 260-263.
Hennewig, U., Schulz, A., Adams, O., Friedrich, W., Gobel, U. & Niehues, T. 2007, "Severe combined immunodeficiency signalized by eosinophilia and lymphopenia in rotavirus infected infants", Klinische Padiatrie, vol. 219, no. 6, pp. 343-347.
Honig, M., Albert, M.H., Schulz, A., Sparber-Sauer, M., Schutz, C., Belohradsky, B., Gungor, T., Rojewski, M.T., Bode, H., Pannicke, U., Lippold, D., Schwarz, K., Debatin, K.M., Hershfield, M.S. & Friedrich, W. 2007, "Patients with adenosine deaminase deficiency surviving after hematopoietic stem cell transplantation are at high risk of CNS complications", Blood, vol. 109, no. 8, pp. 3595-3602.
Horn, B., Viele, M., Mentzer, W., Mogck, N., DeSantes, K. & Cowan, M. 1999, "Autoimmune hemolytic anemia in patients with SCID after T cell-depleted BM and PBSC transplantation", Bone marrow transplantation, vol. 24, no. 9, pp. 1009-1013.
Jones, J.F., Ritenbaugh, C.K., Spence, M.A. & Hayward, A. 1991, "Severe combined immunodeficiency among the Navajo. I. Characterization of phenotypes, epidemiology, and population genetics", Human biology; an international record of research, vol. 63, no. 5, pp. 669-682.
Kane, L., Gennery, A.R., Crooks, B.N., Flood, T.J., Abinun, M. & Cant, A.J. 2001, "Neonatal bone marrow transplantation for severe combined immunodeficiency", Archives of disease in childhood.Fetal and neonatal edition, vol. 85, no. 2, pp. F110-3.
Kline, R.M., Stiehm, E.R. & Cowan, M.J. 1996, "Bone marrow 'boosts' following T cell-depleted haploidentical bone marrow transplantation", Bone marrow transplantation, vol. 17, no. 4, pp. 543-548.
Knutsen, A.P. & Wall, D.A. 2000, "Umbilical cord blood transplantation in severe T-cell immunodeficiency disorders: two-year experience", Journal of clinical immunology, vol. 20, no. 6, pp. 466-476.
Laffort, C., Le Deist, F., Favre, M., Caillat-Zucman, S., Radford-Weiss, I., Debre, M., Fraitag, S., Blanche, S., Cavazzana-Calvo, M., de Saint Basile, G., de Villartay, J.P., Giliani, S., Orth, G., Casanova, J.L., Bodemer, C. & Fischer, A. 2004, "Severe cutaneous papillomavirus disease after haemopoietic stem-cell transplantation in patients with severe combined immune deficiency caused by common gammac cytokine receptor subunit or JAK-3 deficiency", Lancet, vol. 363, no. 9426, pp. 2051-2054.
Mazzolari, E., Forino, C., Guerci, S., Imberti, L., Lanfranchi, A., Porta, F. & Notarangelo, L.D. 2007, "Long-term immune reconstitution and clinical outcome after stem cell transplantation for severe T-cell immunodeficiency", The Journal of allergy and clinical immunology, vol. 120, no. 4, pp. 892-899.
McGhee, S.A., Stiehm, E.R., Cowan, M., Krogstad, P. & McCabe, E.R. 2005, "Two-tiered universal newborn screening strategy for severe combined immunodeficiency", Molecular genetics and metabolism, vol. 86, no. 4, pp. 427-430.
McGhee, S.A., Stiehm, E.R. & McCabe, E.R. 2005, "Potential costs and benefits of newborn screening for severe combined immunodeficiency", The Journal of pediatrics, vol. 147, no. 5, pp. 603-608.
Monafo, W.J., Polmar, S.H., Neudorf, S., Mather, A. & Filipovich, A.H. 1992, "A hereditary immunodeficiency characterized by CD8+ T lymphocyte deficiency and impaired lymphocyte activation", Clinical and experimental immunology, vol. 90, no. 3, pp. 390-393.
Myers, L.A., Patel, D.D., Puck, J.M. & Buckley, R.H. 2002, "Hematopoietic stem cell transplantation for severe combined immunodeficiency in the neonatal period leads to superior thymic output and improved survival", Blood, vol. 99, no. 3, pp. 872-878.
NHS Centre for Reviews and Dissemination March 2001, Accessed: October 17, 2008, Undertaking systematic reviews of research on effectiveness: CRD's guidance for those carrying out or commissioning reviews.
O'Marcaigh, A.S., DeSantes, K., Hu, D., Pabst, H., Horn, B., Li, L. & Cowan, M.J. 2001, "Bone marrow transplantation for T-B- severe combined immunodeficiency disease in Athabascan-speaking native Americans", Bone marrow transplantation, vol. 27, no. 7, pp. 703-709.
Patel, D.D., Gooding, M.E., Parrott, R.E., Curtis, K.M., Haynes, B.F. & Buckley, R.H. 2000, "Thymic function after hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency", The New England journal of medicine, vol. 342, no. 18, pp. 1325-1332.
Pandor, A., Eastham, J., Beverley, C., Chilcott, J. & Paisley, S. 2004, "Clinical effectiveness and cost-effectiveness of neonatal screening for inborn errors of metabolism using tandem mass spectrometry: a systematic review", Health technology assessment (Winchester, England), vol. 8, no. 12, pp. iii, 1-121.
Patel, D.D., Gooding, M.E., Parrott, R.E., Curtis, K.M., Haynes, B.F. & Buckley, R.H. 2000, "Thymic function after hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency", The New England journal of medicine, vol. 342, no. 18, pp. 1325-1332.
Pollitt, R.J., Green, A., McCabe, C.J., Booth, A., Cooper, N.J., Leonard, J.V., Nicholl, J., Nicholson, P., Tunaley, J.R. & Virdi, N.K. 1997, "Neonatal screening for inborn errors of metabolism: cost, yield and outcome", Health technology assessment (Winchester, England), vol. 1, no. 7, pp. i-iv, 1-202.
Puck, J.M. 2007, "Severe combined immunodeficiency: new advances in diagnosis and treatment", Immunologic research, vol. 38, no. 1-3, pp. 64-67.
Puck, J.M. & SCID Newborn Screening Working Group 2007, "Population-based newborn screening for severe combined immunodeficiency: steps toward implementation", The Journal of allergy and clinical immunology, vol. 120, no. 4, pp. 760-768.
Rao, K., Amrolia, P.J., Jones, A., Cale, C.M., Naik, P., King, D., Davies, G.E., Gaspar, H.B. & Veys, P.A. 2005, "Improved survival after unrelated donor bone marrow transplantation in children with primary immunodeficiency using a reduced-intensity conditioning regimen", Blood, vol. 105, no. 2, pp. 879-885.
Rao, K., Amrolia, P.J., Jones, A., Cale, C.M., Naik, P., King, D., Davies, G.E., Gaspar, H.B. & Veys, P.A. 2005, "Improved survival after unrelated donor bone marrow transplantation in children with primary immunodeficiency using a reduced-intensity conditioning regimen", Blood, vol. 105, no. 2, pp. 879-885.
Roberts, J.L., Lengi, A., Brown, S.M., Chen, M., Zhou, Y.J., O'Shea, J.J. & Buckley, R.H. 2004, "Janus kinase 3 (JAK3) deficiency: clinical, immunologic, and molecular analyses of 10 patients and outcomes of stem cell transplantation", Blood, vol. 103, no. 6, pp. 2009-2018.
Schmidt, M., Hacein-Bey-Abina, S., Wissler, M., Carlier, F., Lim, A., Prinz, C., Glimm, H., Andre-Schmutz, I., Hue, C., Garrigue, A., Le Deist, F., Lagresle, C., Fischer, A., Cavazzana-Calvo, M. & von Kalle, C. 2005, "Clonal evidence for the transduction of CD34+ cells with lymphomyeloid differentiation potential and self-renewal capacity in the SCID-X1 gene therapy trial", Blood, vol. 105, no. 7, pp. 2699-2706.
Slatter, M.A., Brigham, K., Dickinson, A.M., Harvey, H.L., Barge, D., Jackson, A., Bown, N., Flood, T.J., Cant, A.J., Abinun, M. & Gennery, A.R. 2008, "Long-term immune reconstitution after anti-CD52-treated or anti-CD34-treated hematopoietic stem cell transplantation for severe T-lymphocyte immunodeficiency", The Journal of allergy and clinical immunology, vol. 121, no. 2, pp. 361-367.
Smogorzewska, E.M., Brooks, J., Annett, G., Kapoor, N., Crooks, G.M., Kohn, D.B., Parkman, R. & Weinberg, K.I. 2000, "T cell depleted haploidentical bone marrow transplantation for the treatment of children with severe combined immunodeficiency", Archivum Immunologiae et Therapiae Experimentalis, vol. 48, no. 2, pp. 111-118.
Stephan, J.L., Vlekova, V., Le Deist, F., Blanche, S., Donadieu, J., De Saint-Basile, G., Durandy, A., Griscelli, C. & Fischer, A. 1993, "Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients", The Journal of pediatrics, vol. 123, no. 4, pp. 564-572.
Stocks, R.M., Thompson, J.W., Church, J.A., Kun, S. & Simms, E. 1999, "Severe combined immunodeficiency: otolaryngological presentation and management", The Annals of Otology, Rhinology, and Laryngology, vol. 108, no. 4, pp. 403-407.
Touraine, J.L., Plotnicky, H., Roncarolo, M.G., Bacchetta, R. & Gebuhrer, L. 2007, "Immunological lessons learnt from patients transplanted with fully mismatched stem cells", Immunologic research, vol. 38, no. 1-3, pp. 201-209.
van Leeuwen, J.E., van Tol, M.J., Joosten, A.M., Schellekens, P.T., van den Bergh, R.L., Waaijer, J.L., Oudeman-Gruber, N.J., van der Weijden-Ragas, C.P., Roos, M.T. & Gerritsen, E.J. 1994, "Relationship between patterns of engraftment in peripheral blood and immune reconstitution after allogeneic bone marrow transplantation for (severe) combined immunodeficiency", Blood, vol. 84, no. 11, pp. 3936-3947.
Veys, P., Rao, K. & Amrolia, P. 2005, "Stem cell transplantation for congenital immunodeficiencies using reduced-intensity conditioning", Bone marrow transplantation, vol. 35 Suppl 1, pp. S45-7.
Vossen, J.M., van Leeuwen, J.E., van Tol, M.J., Joosten, A.M., van den Berg, H., Gerritsen, E.J., Haraldsson, A. & Khan, P.M. 1993, "Chimerism and immune reconstitution following allogeneic bone marrow transplantation for severe combined immunodeficiency disease", Immunodeficiency, vol. 4, no. 1-4, pp. 311-313.
Waruiru, C., Slatter, M.A., Taylor, C., Ramesh, V., Flood, T.J., Abinun, M., Cant, A.J. & Gennery, A.R. 2007, "Outcome of hematopoietic stem cell transplantation in severe combined immune deficiency with central nervous system viral infection", The Pediatric infectious disease journal, vol. 26, no. 2, pp. 129-133.

Wijnaendts, L., Le Deist, F., Griscelli, C. & Fischer, A. 1989, "Development of immunologic functions after bone marrow transplantation in 33 patients with severe combined immunodeficiency", Blood, vol. 74, no. 6, pp. 2212-2219.

 

_________________________________________________________________________

Version:  11/12/08
Authors:
Ellen Lipstein, Alixandra Knapp, James Perrin

Evidence Review Group:  Chairperson, James M. Perrin, MD
(MGH Center for Child and Adolescent Health Policy)

 

Committee Members:


Marsha Browning, MD, MPH, MMSc. (Massachusetts General Hospital)

Jennifer DeZarn, BA
(MGH Center for Child and Adolescent Health Policy)

Nancy Green, MD
(Columbia University)

Alex R. Kemper, MD, MPH, MS
(Duke University)

Alixandra Knapp, MS
(MGH Center for Child and Adolescent Health Policy)

Ellen Lipstein, MD
(MGH Center for Child and Adolescent Health Policy)

Lisa Prosser, PhD
(University of Michigan)

Denise Queally, JD
(Consumer Representative)

Sienna Vorono, BA
(MGH Center for Child and Adolescent Health Policy; Brown Medical School)


This review was made possible by subcontract number SC-07-028 to Massachusetts General Hospital, Center for Child and Adolescent Health Policy under prime contract number HHSP23320045014XI to Altarum Institute, from the Maternal and Child Health Bureau (MCHB) (Title V, Social Security Act), Health Resources and Services Administration (HRSA), U.S. Department of Health and Human Services (DHHS).