Adenosine deaminase (ADA) deficiency is a systemic metabolic disease that causes an autosomal recessive variant of severe combined immunodeficiency (SCID) and less consistently other complications including neurologic abnormalities. Hematopoietic stem cell transplantation (HSCT) is able to correct the immunodeficiency, whereas control of nonimmunologic complications has not been extensively explored. We applied HSCT in 15 ADA-deficient patients consecutively treated at our institutions since 1982 and analyzed long-term outcome. Seven patients received transplants without conditioning from HLA-matched family donors (MFDs); the other 8 patients received conditioning and were given transplants either from HLA-mismatched family donors (MMFDs; n = 6) or from matched unrelated donors (MUDs; n = 2). At a mean follow-up period of 12 years (range, 4-22 years), 12 patients are alive with stable and complete immune reconstitution (7 of 7 after MFD, 4 of 6 after MMFD, and 1 of 2 after MUD transplantation). Six of 12 surviving patients show marked neurologic abnormalities, which include mental retardation, motor dysfunction, and sensorineural hearing deficit. We were unable to identify disease or transplantation-related factors correlating with this divergent neurologic outcome. The high rate of neurologic abnormalities observed in long-term surviving patients with ADA deficiency indicates that HSCT commonly fails to control CNS complications in this metabolic disease.

One cause of autosomal recessive severe combined immunodeficiency (SCID) is a deficiency of the enzyme adenosine deaminase (ADA).1,2  ADA is an ubiquitously expressed enzyme involved in the degradation of adenosine and deoxyadenosine. In ADA-deficient patients these nucleosides and their phosphorylated metabolites accumulate, which, by mechanisms not yet completely clarified, induce a profound impairment in particular of the lymphoid system.2,3  Affected infants usually present early in life with life-threatening infections and the prognosis is extremely poor unless the immunodeficiency is treated. Less severe forms of ADA deficiency have been described and are characterized by a delayed onset of symptoms and initially less profound immunologic abnormalities.2 

Beside the immunodeficiency, other clinical findings may be encountered in ADA-deficient patients, including skeletal abnormalities and rarely also neurologic complications. In an analysis of 117 SCID patients, developmental delay was reported in 6 of 16 ADA-deficient patients as a disease-specific finding.4  In another report, 3 of 23 ADA-deficient patients suffered from cortical blindness and pyramidal and extrapyramidal motor dysfunction.5  In a further report, 2 siblings with ADA deficiency were found to have sensorineural hearing deficit.6  Because complications from infections usually predominate in the clinical presentation of infants with ADA deficiency, the full spectrum of nonimmunologic manifestations of the disorder and their natural course may be obscured.

As in patients with nonmetabolic forms of SCID, hematopoietic stem cell transplantation (HSCT) is able to correct the immunodeficiency in ADA deficiency. After HLA-identical HSCT, excellent results have been reported with survival rates approaching 100%.4,7,8  Exploration of HLA-mismatched HSCT in ADA deficiency has provided less favorable results compared to those obtained in other variants of SCID, mainly due to a high rate of graft failures.4,9-11  By the use of cytoreductive conditioning, this difficulty was overcome.8,12  Therapeutic alternatives in ADA deficiency are long-term enzyme replacement therapy with polyethylene glycol-ADA (PEG-ADA)13,14  as well as more recently introduced successful gene therapy.15-17 

We analyzed cumulative experience of HSCT in 15 ADA-deficient patients treated at 2 centers and report a high rate of marked neurologic abnormalities in long-term surviving patients.

Approval was obtained from the Institutional Review Board of the University of Ulm for these studies. Informed consent was obtained in accordance with the Declaration of Helsinki.

Patients

Within a group of 170 patients with SCID diagnosed at our institutions since 1982, 15 patients suffered from ADA deficiency and were treated by HSCT (13 at Children's Hospital in Ulm, 2 at Children's Hospital in Munich). These patients were of different origin (5 German, 4 Turkish, 3 Swiss, 1 Afghan, 1 Albanian, 1 Austrian). Five patients were born to consanguineous parents (Table 4). Two patients were identical twins. The diagnosis of ADA deficiency was based on enzyme activity in erythrocytes, which was undetectable in 14 and markedly reduced in one case.

HSCT

In 7 cases HLA-matched family donors (MFDs) were available. These were either siblings (n = 5) or HLA-genotypically identical parents (n = 2). In 6 cases, donors were HLA-haploidentical parents (mismatched family donors, MMFDs) and in 2 cases matched unrelated donors (MUDs). Two patients had received prior enzyme replacement therapy with PEG-ADA (UPN U322 for 3 weeks, U457 for 9 months), which was discontinued several weeks before transplantation (Table 2). Patients received bone marrow except for 3 patients who received peripheral-blood stem cells (PBSCs) harvested after G-CSF treatment of donors (Table 2). Transplants from MMFD and from one MUD (UPN U442) were depleted of T cells to prevent graft versus host disease (GvHD), using soybean lectin agglutination and E-rosette formation or positive selection of CD34+ cells for PBSCs. Recipients of T-cell–depleted transplants received no further GvHD prophylaxis. Regimens used in the other patients are listed in Table 2. Eight patients underwent pretransplantation conditioning, which consisted of a combination of busulfan and cyclophosphamide with some variation in the dosage of busulfan (Table 2 lists details).

Table 1

Clinical and immunologic findings at diagnosis in 15 ADA-deficient patients

UPNSexAgeClinical presentation/infectionT cells/mm3B cells/mm3NK cells/mm3Eos/mm3IgG, g/LIgA, g/LIgM, gL
U466 1 mo Pneumonia (RSV), diarrhea 102 420 4.8 ND ND 
U396 3 mo Pneumonia (RSV), mechanical ventilation 120 3.7 ND ND 
U301 2 wk Pneumonia (RSV) 17 85 79 1500 2.1 ND ND 
U175 2 mo Diarrhea, FTT 60 30 1300 1.8 ND ND 
U118 2 mo Lymphadenopathy after BCG-vaccination Not done 40 1.4 0.1 0.4 
M1052 4 mo Recurrent pyodermia, FTT 20 50 125 150 0.9 ND 0.05 
M1053 4 mo Diarrhea, FTT 135 105 0.9 ND 0.05 
U442 3 mo Diarrhea, FTT 70 40 1000 5.4 0.25 0.21 
U457 12 mo Bronchitis, autoimmune thyroiditis 690 50 650 17.0 0.43 0.36 
U444 Birth None 12 250 6.9 ND 0.07 
U322 2 wk Pneumonia, mechanical ventilation, FTT 820 3.4 ND ND 
U66 Birth UTI, PCP 10 1500 4.6 ND ND 
U51 2 wk Lymphadenopathy after BCG-vaccination, pneumonia, FTT 200 55 Not done 1350 2.7 ND ND 
U36 1 mo Pneumonia, diarrhea, FTT 200 Not done 750 4.4 ND ND 
U26 1 mo Lymphadenopathy after BCG-vaccination, pneumonia, sepsis/meningitis, diarrhea 65 70 Not done 170 8.0 ND ND 
UPNSexAgeClinical presentation/infectionT cells/mm3B cells/mm3NK cells/mm3Eos/mm3IgG, g/LIgA, g/LIgM, gL
U466 1 mo Pneumonia (RSV), diarrhea 102 420 4.8 ND ND 
U396 3 mo Pneumonia (RSV), mechanical ventilation 120 3.7 ND ND 
U301 2 wk Pneumonia (RSV) 17 85 79 1500 2.1 ND ND 
U175 2 mo Diarrhea, FTT 60 30 1300 1.8 ND ND 
U118 2 mo Lymphadenopathy after BCG-vaccination Not done 40 1.4 0.1 0.4 
M1052 4 mo Recurrent pyodermia, FTT 20 50 125 150 0.9 ND 0.05 
M1053 4 mo Diarrhea, FTT 135 105 0.9 ND 0.05 
U442 3 mo Diarrhea, FTT 70 40 1000 5.4 0.25 0.21 
U457 12 mo Bronchitis, autoimmune thyroiditis 690 50 650 17.0 0.43 0.36 
U444 Birth None 12 250 6.9 ND 0.07 
U322 2 wk Pneumonia, mechanical ventilation, FTT 820 3.4 ND ND 
U66 Birth UTI, PCP 10 1500 4.6 ND ND 
U51 2 wk Lymphadenopathy after BCG-vaccination, pneumonia, FTT 200 55 Not done 1350 2.7 ND ND 
U36 1 mo Pneumonia, diarrhea, FTT 200 Not done 750 4.4 ND ND 
U26 1 mo Lymphadenopathy after BCG-vaccination, pneumonia, sepsis/meningitis, diarrhea 65 70 Not done 170 8.0 ND ND 

IgG levels are given before substitution.

Eos indicates eosinophilic granulocytes; U proceeding UPN numbers indicates patients treated in Ulm, M patients treated in Munich; m, male; f, female; RSV, respiratory syncytial virus; ND, not detectable; FTT, failure to thrive; BCG, Bacille Calmette Guérin; UTI, urinary tract infection; PCP, Pneumocystis jirovecii pneumonia.

Table 2

Details of transplantation

UPNAge at HSCTDonorMismatch (HLA-A, -B, -DR)GraftT-cell depletionConditioningBu dosage, mg/kgGvHD prophylaxisGvHD grade II or higher, acute/chronicOutcome
U466 2 mo MSD BM No None No No No/no Alive 
U396 3 mo MFD BM No None No No No/no Alive 
U301 2 mo MFD BM No None No CSA No/no Alive 
U175 3 mo MSD BM No None No PDN II/no Alive 
U118 3 mo MSD BM No None No CSA No/no Alive 
M1052 5 mo MSD BM No None No No No/no Alive 
M1053 5 mo MSD BM No None No No No/no Alive 
U442 1 y 5 mo MUD PBSC Yes Bu, Cy 13 No No/no Alive 
U457 2 y 6 mo MUD BM No Bu, Cy 16 CSA, MTX No/no Death day +61, CMV, adenovirus 
U444 2 mo MMFD PBSC Yes Bu, Cy 13 No No/no Alive 
U322 4 mo MMFD PBSC Yes Bu, Cy No No/no Death day +34, aspergillosis 
U66 2 mo MMFD BM Yes Bu, Cy No III/No Death day +23, aspergillosis 
U51 6 mo MMFD BM Yes Bu, Cy No II/yes Alive 
U36 4 mo MMFD BM Yes Bu, Cy No No/no Alive 
U26 8 mo MMFD BM Yes Bu, Cy 12 No No/no Alive 
UPNAge at HSCTDonorMismatch (HLA-A, -B, -DR)GraftT-cell depletionConditioningBu dosage, mg/kgGvHD prophylaxisGvHD grade II or higher, acute/chronicOutcome
U466 2 mo MSD BM No None No No No/no Alive 
U396 3 mo MFD BM No None No No No/no Alive 
U301 2 mo MFD BM No None No CSA No/no Alive 
U175 3 mo MSD BM No None No PDN II/no Alive 
U118 3 mo MSD BM No None No CSA No/no Alive 
M1052 5 mo MSD BM No None No No No/no Alive 
M1053 5 mo MSD BM No None No No No/no Alive 
U442 1 y 5 mo MUD PBSC Yes Bu, Cy 13 No No/no Alive 
U457 2 y 6 mo MUD BM No Bu, Cy 16 CSA, MTX No/no Death day +61, CMV, adenovirus 
U444 2 mo MMFD PBSC Yes Bu, Cy 13 No No/no Alive 
U322 4 mo MMFD PBSC Yes Bu, Cy No No/no Death day +34, aspergillosis 
U66 2 mo MMFD BM Yes Bu, Cy No III/No Death day +23, aspergillosis 
U51 6 mo MMFD BM Yes Bu, Cy No II/yes Alive 
U36 4 mo MMFD BM Yes Bu, Cy No No/no Alive 
U26 8 mo MMFD BM Yes Bu, Cy 12 No No/no Alive 

Bu indicates busulfan; MSD, matched sibling donor; BM, bone marrow; CSA, cyclosporine A; PDN, prednisolone; PBSC, peripheral blood stem cells; Cy, cyclophosphamide (4 × 50 mg/kg); MTX, methotrexate; and CMV, cytomegalovirus.

Prior to treatment, parents gave written informed consent for the treatment procedures and for anonymous scientific evaluation.

Immunologic reconstitution and chimerism

Lymphocyte phenotyping was performed by fluorescence-activated cell sorting (FACS) analysis using commercially available monoclonal antibodies. T-cell function was determined by standard proliferative assays as previously described.18  Chimerism of white blood cells was analyzed by HLA typing, fluorescence in situ hybridization (FISH), or short tandem repeat (STR) analysis. Mixed chimerism was analyzed in more detail. For this, T cells (CD3+), B cells (CD19+), and monocytes (CD14+) were isolated by FACS (FACSAria, Becton Dickinson, Heidelberg, Germany) resulting in more than 98% purity of cell populations. Erythrocyte chimerism was assessed by blood group determination. After normalization of immune functions, patients were discharged home and were usually re-evaluated once yearly at our centers.

Posttransplantation levels of erythrocyte ADA activity and adenine nucleotides

Posttransplantation ADA activity in red blood cells was determined by a radiochemical thin-layer chromatographic method (conversion of [14C]adenosine to inosine) at Duke University Medical Center (Durham, NC) and in 3 patients at the Children's Hospital (Zurich, Switzerland). Intracellular levels of deoxyadenosine-X-phosphate (dAXP) and adenosine-X-phosphate (AXP) were determined by high-pressure liquid chromatography from samples of packed red cells collected at the most recent presentation of the patients.19,20 

Sequencing of the ADA gene

Coding sequences and the exon/intron boundaries of the ADA gene were amplified using the Taq polymerase system (Hot Start, Qiagen, Hilden, Germany) from DNA originating from 13 individuals from 13 families. Primers used for polymerase chain reaction (PCR) and sequencing are available on request.

Clinical presentation before HSCT

Most patients (14 of 15) presented within the first 4 months of life with respiratory tract infections of viral or bacterial origin, with intractable diarrhea and failure to thrive (Table 1). Diagnosis in 2 infants was made immediately after birth because of positive family histories. During the first months of life, none of the patients had unusual neurologic findings. One patient developed bacterial meningitis at 6 months of age, which was treated successfully. One patient (UPN U457) had a less severe form of ADA deficiency with only minor infections during the first year of life. With the exception of this patient, all other patients had absent or extremely low numbers of T, B and natural killer (NK) cells and thus findings characteristic of severe ADA deficiency. If detectable, T cells were autologous and not of maternal origin as demonstrated by HLA-typing or XX/XY-FISH analysis. HSCT was performed within the first 6 months of life except in 3 children, of whom 2 were older than 1 year.

Outcome after transplantation from HLA-identical family donors

After HLA-identical family donor transplantation all 7 patients survive after a mean follow-up time of 9.5 years (range, 3.9-13.5 years). These patients continue to show complete immune reconstitution, with T-cell counts ranging from 1000 to 2300/μL and with regular in vitro T-cell proliferative responses. B-cell numbers are low in 5 of 7 patients; however, immunoglobulin serum levels are normal without substitution and all patients developed positive antibody responses after vaccination (Table 3). Chimerism analysis shows T cells to be exclusively of donor origin. Analysis of B cells and monocytes reveals mixed chimerism with variable proportions of donor and host cells (Table 4). In 4 of 7 cases donors and recipients had different blood groups, and in all 4 cases mixed red-cell chimerism was observed.

Table 3

Immunologic reconstitution

UPNCurrent ageT-cell reconstitution
B-cell reconstitution
CD3+/mm3CD4+/mm3CD8+/mm3PHA, SICD19+/mm3IVIGAntibody responses
TetanusDiphtheriaOther
U466 3 y 11 mo 1170 510 560 110 120 No HiB 
U396 6 y 1 mo 2325 1150 790 450 190 No ND 
U301 8 y 10 mo 1180 450 330 760 100 No ND 
U175 13 y 6 mo 1010 470 350 210 175 No HiB 
U118 17 y 1 mo 1090 700 390 190 85 No ND 
M1052 8 y 8 mo 1625 1070 325 350 510 No Pneu, HiB 
M1053 8 y 8 mo 1720 1060 345 300 580 No Pneu, HiB 
U442 4 y 7 mo 1580 850 500 490 470 No ND 
U444 4 y 7 mo 2060 1200 640 430 1050 No ND 
U51 20 y 10 mo 2690 1500 1390 360 910 No ND 
U36 21 y 10 mo 1730 870 680 1120 260 No ND 
U26 22 y 2 mo 1570 850 480 2350 200 No Pneu, HiB 
Reference value > 5 y 700-4200 300-2100 200-1800 > 100 100-1600 — — — — 
UPNCurrent ageT-cell reconstitution
B-cell reconstitution
CD3+/mm3CD4+/mm3CD8+/mm3PHA, SICD19+/mm3IVIGAntibody responses
TetanusDiphtheriaOther
U466 3 y 11 mo 1170 510 560 110 120 No HiB 
U396 6 y 1 mo 2325 1150 790 450 190 No ND 
U301 8 y 10 mo 1180 450 330 760 100 No ND 
U175 13 y 6 mo 1010 470 350 210 175 No HiB 
U118 17 y 1 mo 1090 700 390 190 85 No ND 
M1052 8 y 8 mo 1625 1070 325 350 510 No Pneu, HiB 
M1053 8 y 8 mo 1720 1060 345 300 580 No Pneu, HiB 
U442 4 y 7 mo 1580 850 500 490 470 No ND 
U444 4 y 7 mo 2060 1200 640 430 1050 No ND 
U51 20 y 10 mo 2690 1500 1390 360 910 No ND 
U36 21 y 10 mo 1730 870 680 1120 260 No ND 
U26 22 y 2 mo 1570 850 480 2350 200 No Pneu, HiB 
Reference value > 5 y 700-4200 300-2100 200-1800 > 100 100-1600 — — — — 

Reference values for lymphocyte subpopulations are adapted from Comans-Bitter et al.21 

PHA indicates phytohemagglutinin; SI, stimulation index; HiB, Haemophilus influenzae type B; ND, not determined; and Pneu, pneumococcus.

Table 4

Chimerism, metabolic evaluation after HSCT, and ADA genotype

UPNMNCChimerism
Metabolic evaluation after HSCT
Neurologic symptomsConsanguinityADA genotype and predicted effects on protein
T cellB cellMonocytesRed cells, % donorADA activity in red cells, μmol/mg proteindAXP in red cells, nmol/mL RBC
U466 — Dr NA 0.0 34 No Yes c.164_172del9; p.Pro55_Thr57del; homo 
U396 — dr NA 10 U/L (260-650) — Yes Yes c.302G>A; p.Arg101Gln; homo 
U301 — Dr dR NA 0 U/L (260-650) — Yes Yes c.955_959del5; p.Glu319fsX337; homo 
U175 — Dr 0.0 31 No No c.424C>T; p.Arg142X; homo 
U118 —   20 3.2 22 No No c.[646G>A]+[911T>G]; p.[Gly216Arg]+[Leu304Arg] 
M1052 — dr 45 4.1 18 Yes No c.[646G>A]+[955_959del5]; p.[Gly216Arg]+[Glu319fsX337] 
M1053 — dr 45 4.0 16 Yes No c.[646G>A]+[955_959del5]; p.[Gly216Arg]+[Glu319fsX337] 
U442 — — — 100 36.4 No No c.[43C>G]+[466C>T]; p.[His15Asp]+[Arg156Cys] 
U457 — — — — — — — — No c.385G>A;p.Val129Met; homo 
U444 — — — 100 16.5 No Yes c.736C>T; p.Gln246X; homo 
U322 — — — — — — — — Yes c.295G>T; p.Glu99X; homo 
U66 — — — — — — — — No Affected sister of U26 
U51 — — — 100 187 U/L (260-650) — Yes No — 
U36 — — — 80 21.5 No No c.646G>A; p.Gly216Arg; homo 
U26 — — — 100 8.5 Yes No c.646G>A; p.Gly216Arg; homo 
Normal reference — — — — — 63 ± 41 unless indicated otherwise* < 2  — — — 
UPNMNCChimerism
Metabolic evaluation after HSCT
Neurologic symptomsConsanguinityADA genotype and predicted effects on protein
T cellB cellMonocytesRed cells, % donorADA activity in red cells, μmol/mg proteindAXP in red cells, nmol/mL RBC
U466 — Dr NA 0.0 34 No Yes c.164_172del9; p.Pro55_Thr57del; homo 
U396 — dr NA 10 U/L (260-650) — Yes Yes c.302G>A; p.Arg101Gln; homo 
U301 — Dr dR NA 0 U/L (260-650) — Yes Yes c.955_959del5; p.Glu319fsX337; homo 
U175 — Dr 0.0 31 No No c.424C>T; p.Arg142X; homo 
U118 —   20 3.2 22 No No c.[646G>A]+[911T>G]; p.[Gly216Arg]+[Leu304Arg] 
M1052 — dr 45 4.1 18 Yes No c.[646G>A]+[955_959del5]; p.[Gly216Arg]+[Glu319fsX337] 
M1053 — dr 45 4.0 16 Yes No c.[646G>A]+[955_959del5]; p.[Gly216Arg]+[Glu319fsX337] 
U442 — — — 100 36.4 No No c.[43C>G]+[466C>T]; p.[His15Asp]+[Arg156Cys] 
U457 — — — — — — — — No c.385G>A;p.Val129Met; homo 
U444 — — — 100 16.5 No Yes c.736C>T; p.Gln246X; homo 
U322 — — — — — — — — Yes c.295G>T; p.Glu99X; homo 
U66 — — — — — — — — No Affected sister of U26 
U51 — — — 100 187 U/L (260-650) — Yes No — 
U36 — — — 80 21.5 No No c.646G>A; p.Gly216Arg; homo 
U26 — — — 100 8.5 Yes No c.646G>A; p.Gly216Arg; homo 
Normal reference — — — — — 63 ± 41 unless indicated otherwise* < 2  — — — 

Capital letters in “chimerism” columns indicate dominant part of either donor or recipient.

D indicates donor; r, recipient; —, not determined; NA, not applicable (blood groups of donor and recipient identical in ABO and Rh).

*

ADA activity was measured in 2 different laboratories, results are given in μmol/mg protein (M. Hershfield, Durham, NC) or U/L red cells (University Children's Hospital Zurich, Switzerland).

Deceased.

Non–T-cell: mix.

Outcome after MMFD and MUD transplantation

Six patients received transplants from MMFDs and 2 patients from MUDs. In these cases we used cytoreductive conditioning prior to HSCT with the exception of 2 cases, who initially underwent transplantation without conditioning: UPN U26 from his haploidentical father (this case has been previously reported18 ) and U457 from a MUD. Transplanted cells in both cases failed to engraft. Subsequent transplants following conditioning were successful. Causes of death in 3 patients were early infections, associated with acute GvHD in one case. A second patient (UPN U51) developed acute and chronic GvHD, which resolved completely.

The overall survival rate in the 8 patients is 63% (MMFD 4 of 6; MUD 1 of 2), and after a mean follow-up of 14.6 years (range, 4.6-22.2 years), all 5 long-term surviving patients show stable and complete immune reconstitution. Chimerism analysis reveals all blood cells including red cells to be of donor origin, except in one patient who had a small proportion of autologous red cells.

ADA molecular analysis

Molecular analysis of the ADA gene was performed in 13 cases and revealed 2 deletions, 1 nonsense mutation, and 8 missense mutations (Table 4). The missense mutations have all been described previously22-24  and all except one have been tested for residual ADA activity in Escherichia coli.22  For the homozygous missense mutation found in patient U457 with delayed onset of disease, some residual ADA activity would be predicted (class II according to Arredondo-Vega et al22 ). All other missense mutations would be classified as class I (minimal or no residual ADA activity). The genotype, therefore, is in high concordance with the clinical presentation with early onset of disease in all but one patient.

Erythrocyte ADA activity and dAXP levels after transplantation

ADA activity in red cells after HSCT was highest in conditioned patients who developed complete donor red-cell chimerism (Table 4). After HSCT without conditioning, 4 of 6 patients showed detectable levels of ADA activity, which, however, were much lower as compared to conditioned patients. Red-cell dAXP levels were variable, ranging from 2.5- to 17-fold of the upper normal limit (2 nmol/mL packed RBCs; Table 4). These are markedly lower than dAXP levels in red cells of untreated ADA-deficient SCID patients which may approach 2000 nmol/mL.25 

Neurologic outcome after HSCT

Six of the 12 long-term survivors are in excellent health and have no clinical complications, whereas 6 patients suffer from significant neurologic and cognitive deficits, as summarized in Table 5. These patients have learning disabilities, precluding attendance of regular (pre-) school, and require continuous special support. In addition, 4 patients show persistently abnormal gait and 5 patients exhibit a sensorineural hearing deficit. Furthermore, in 4 cases hyperactivity is a prominent problem. Common to all 6 patients was a marked delay in reaching developmental milestones, which usually became obvious during the second year of life. Remarkably, in none of the patients did we observe progressive deterioration or loss of acquired neurologic abilities during later childhood and adolescence.

Table 5

CNS findings in long-term survivors

UPNCurrent ageMajor findingsCNS imaging after HSCT (age)Abnormal findings
Special school
HyperactivityMotor dysfunctionVerbal expressionHearing deficitSeizuresLearning disability
U466 4 y Normal ND − − − − − — − 
U396 6 y Muscular hypotonia (4 mo); sitting without support (18 mo); walking (24 mo); current: gross motor coordination disorder with tiptoe walking, reduced verbal expression (5-10 words), severe learning disability, hyperactivity, continuous special care US: normal (6 mo) − − Severe 
U301 9 y Pendular nystagmus (5 mo); sitting without support (16 mo); walking (24 mo); current: gross motor coordination disorder with tiptoe walking, pendular nystagmus, reduced verbal expression, sensorineural hearing deficit, severe learning disability, hyperactivity, continuous special care US: normal (3 mo) − Severe 
U175 14 y Mild learning disability ND − − − Mild 
U118 17 y Normal, finished regular school CT scan: normal (6 mo) − − − − − — − 
M1052 9 y Developmental delay (4 mo); sitting without support (11 mo); walking (18 mo); current: fine motor and coordination deficit, slurred speech, hyperactivity, mild learning disability ND − Mild 
M1053 9 y Muscular hypotonia (4 mo); sitting without support (18 mo); walking (24 mo); seizures (35 y); current: spastic diplegia, slurred speech, reduced verbal expression (< 10 words), severe learning disability, seizures, continuous special care MRI: mild expansion of external CSF space (6 y) − − Severe 
U442 5 y Normal ND − − − − − — − 
U444 5 y Normal US: normal (3 mo) − − − − − — − 
U51 21 y Hearing deficit (12 mo); sitting without support (12 mo); walking (23 mo); current: reduce expressive speech, attention deficit, moderate to severe learning disability, hyperactivity, needs support in everyday life CT-scan: normal (8 mo+5 y) − Moderate/severe 
U36 22 y Normal, finished regular school ND − − − − − — − 
U26 22 y Generalized muscular hypertonia (11 mo); sitting without support (15 mo); walking (20 mo); current: fine motor and coordination deficit, slurred speech, attention deficit, moderate learning disability, needs support in everyday life ND − − Moderate 
UPNCurrent ageMajor findingsCNS imaging after HSCT (age)Abnormal findings
Special school
HyperactivityMotor dysfunctionVerbal expressionHearing deficitSeizuresLearning disability
U466 4 y Normal ND − − − − − — − 
U396 6 y Muscular hypotonia (4 mo); sitting without support (18 mo); walking (24 mo); current: gross motor coordination disorder with tiptoe walking, reduced verbal expression (5-10 words), severe learning disability, hyperactivity, continuous special care US: normal (6 mo) − − Severe 
U301 9 y Pendular nystagmus (5 mo); sitting without support (16 mo); walking (24 mo); current: gross motor coordination disorder with tiptoe walking, pendular nystagmus, reduced verbal expression, sensorineural hearing deficit, severe learning disability, hyperactivity, continuous special care US: normal (3 mo) − Severe 
U175 14 y Mild learning disability ND − − − Mild 
U118 17 y Normal, finished regular school CT scan: normal (6 mo) − − − − − — − 
M1052 9 y Developmental delay (4 mo); sitting without support (11 mo); walking (18 mo); current: fine motor and coordination deficit, slurred speech, hyperactivity, mild learning disability ND − Mild 
M1053 9 y Muscular hypotonia (4 mo); sitting without support (18 mo); walking (24 mo); seizures (35 y); current: spastic diplegia, slurred speech, reduced verbal expression (< 10 words), severe learning disability, seizures, continuous special care MRI: mild expansion of external CSF space (6 y) − − Severe 
U442 5 y Normal ND − − − − − — − 
U444 5 y Normal US: normal (3 mo) − − − − − — − 
U51 21 y Hearing deficit (12 mo); sitting without support (12 mo); walking (23 mo); current: reduce expressive speech, attention deficit, moderate to severe learning disability, hyperactivity, needs support in everyday life CT-scan: normal (8 mo+5 y) − Moderate/severe 
U36 22 y Normal, finished regular school ND − − − − − — − 
U26 22 y Generalized muscular hypertonia (11 mo); sitting without support (15 mo); walking (20 mo); current: fine motor and coordination deficit, slurred speech, attention deficit, moderate learning disability, needs support in everyday life ND − − Moderate 

US indicates ultrasound; CSF, cerebrospinal fluid; +, present finding; and −, absent finding.

Most patients had cranial ultrasound studies, magnetic resonance imaging (MRI) or computed tomography (CT) scans before transplantation and at follow-up evaluations. With the exception of one patient these were normal (Table 5). In the exceptional case, who had suffered from bacterial meningitis prior to successful transplantation, nonspecific bilateral frontal and parietal calcifications were noted.

Of particular interest was our experience in the identical twins (UPN M1052 and M1053). They underwent transplantation simultaneously from a healthy HLA-identical sister without conditioning. There were no major differences in medication or infections in the pretransplantation and posttransplantation period. One twin shows a distinctly more severe learning disability and is suffering from epilepsy with only partial response to anticonvulsive therapy. Her sister is less severely affected and has never experienced seizures.

We analyzed various parameters in the 12 surviving patients to recognize possible correlations with neurologic outcome. All infants had an uneventful perinatal and neonatal history. CNS infections were neither diagnosed nor suspected, except in the one case mentioned with bacterial meningitis, from which he recovered. All survivors had severe forms of ADA deficiency with early onset of infections and complete absence of enzyme activity in red cells. No evidence was found for a correlation of neurologic outcome with parameters such as genotype of the disease, age at transplantation, use of conditioning prior to transplantation, donor type or donor-cell chimerism, posttransplantation dAXP, levels and serious complications such as mechanical ventilation for respiratory failure (Tables 1 and 6).

Table 6

Comparison of patients with and without CNS symptoms

No.CNS+CNS−
Consanguineous parents 
Nonconsanguineous parents 
Age at transplantation younger than 6 mo 10 
Age at transplantation older than 6 mo 
Transplantation with conditioning 
Transplantation without conditioning 
Complete chimerism 
Incomplete chimerism 
Hospitalization less than 5 mo 
Hospitalization more than 5 mo 
Current age younger than 10 y 
Current age older than 10 y 
dAXP levels in red cells after HSCT elevated 
dAXP levels in red cells after HSCT normal 
No.CNS+CNS−
Consanguineous parents 
Nonconsanguineous parents 
Age at transplantation younger than 6 mo 10 
Age at transplantation older than 6 mo 
Transplantation with conditioning 
Transplantation without conditioning 
Complete chimerism 
Incomplete chimerism 
Hospitalization less than 5 mo 
Hospitalization more than 5 mo 
Current age younger than 10 y 
Current age older than 10 y 
dAXP levels in red cells after HSCT elevated 
dAXP levels in red cells after HSCT normal 

CNS+ indicates presence of clinically evident neurologic symptoms; and CNS−, absence of neurologic symptoms.

In ADA deficiency, transplantation of hematopoietic stem cells serves 2 purposes. One is the establishment of a functioning immune system by donor lymphocytes, similar as in other SCID variants. The other is the improvement of metabolic abnormalities by the enzymatic activity provided by donor cells. The latter mechanism may be important, in particular, for the control of nonimmunologic manifestations associated with this systemic metabolic disease.

In the study presented here we evaluated the outcome of HSCT in 15 SCID patients with ADA deficiency. Twelve patients are alive with complete reconstitution of immune functions after a mean follow-up period of 12 years (range, 4-22 years). Following HLA-identical family donor transplantation the survival rate is 100%, confirming similar good results reported previously.4,7,8  In these patients, who underwent the procedure without conditioning, donor-cell engraftment is incomplete and restricted mainly to T cells, whereas B cells and other blood cells remain predominantly of host type. This is in contrast to patients receiving a transplant after conditioning, which always resulted in complete donor-cell chimerism. It is interesting to note that humoral immunity is normal in the former group of patients, despite mixed B-cell chimerism and lower absolute numbers of B cells compared to conditioned patients (Table 3). The contribution of autologous B cells to humoral immune reconstitution after HSCT in ADA deficiency remains an open question.

As also reported by others, transplantation of T-cell–depleted, HLA-haploidentical stem cells in ADA-deficient patients carries a substantial risk for complete graft failure. This is in contrast to the experience in nonmetabolic variants of SCID where donor T cells commonly develop in the absence of conditioning. The basis for this discrepancy remains unclear. One hypothesis is that ADA enzyme activity provided by the transplant enhances the potential of residual host lymphocytes to resist engraftment of allogeneic donor cells. This hypothesis is supported by the successful engraftment after conditioning observed in 2 of our patients who formerly had rejected their grafts. In our series 4 of 6 patients are alive with normal immunity after HLA haploidentical, parental donor transplantation. Nevertheless, the need for conditioning represents a major limitation to this approach due to the risk of developing toxic complications. An alternative treatment in ADA deficiency represents long-term enzyme replacement therapy with PEG-ADA by regular intramuscular injections.2,26,27  This therapy has been effective in restoring cellular and humoral immunity in the majority of treated patients. Significant disadvantages are high costs and the necessity for lifelong application of this therapy. Moreover, recent studies indicate that immunologic functions may decrease after several years.28,29  Recently, gene therapy has been introduced with success in the treatment of ADA deficiency.15-17  Results are promising but experience is confined to a very limited number of patients and longer follow-up is required to evaluate the full potential as well as possible risks of this new treatment.

A major finding in our study is a high rate of neurologic abnormalities that we observed in 6 of 12 long-term surviving patients. These patients became symptomatic during the first 2 years of life and suffer from significant learning disabilities as well as other neurologic abnormalities including sensorineural hearing deficit, muscular hypertonia, and problems with coordination and expressive speech. Based on our experience in approximately 100 long-term surviving patients undergoing transplantation for nonmetabolic variants of SCID, we have no evidence that the transplantation procedure as applied in this disorder carries a significant risk to induce CNS complications. There are only 2 other patients, both suffering from SCID of unknown etiology, who show unexplained mental retardation after HSCT. We previously reported sensorineural hearing defects in patients with SCID and reticular dysgenesis as an isolated finding not associated with mental retardation or other neurologic findings.30 

In the present study we obtained no evidence for a correlation of neurologic abnormalities with complications before or after HSCT or with variables of the transplantation procedure such as the use of conditioning, donor type, or length of hospitalization. Our findings thus would indicate that CNS abnormalities observed in ADA-deficient patients most likely are caused by metabolic abnormalities of the underlying disorder rather than representing complications of the treatment procedure. One other previous study addressed neurologic outcome after HSCT in ADA-deficient patients.31  In this study, which included 16 patients as well as a control group given transplants for nonmetabolic SCID, the authors reported striking behavioral abnormalities in ADA-deficient patients. In contrast to our findings an evaluation of motor function did not reveal abnormalities. Interestingly, in ADA-deficient patients the authors observed an inverse correlation of cognitive function with toxin levels before transplantation. In our patients no correlation to genotype or posttransplantation dAXP levels could be demonstrated. Unfortunately, no samples were available for determination of dAXP levels before transplantation. However, indirect parameters such as early age at clinical presentation, pretransplantation red-cell ADA activity, and severe genotypes indicate a homogeneous composition of our group in this regard.

CNS abnormalities including motor dysfunction have been described in several ADA-deficient patients at diagnosis and before the initiation of any therapy.5,32,33  Hirschhorn et al postulated that these abnormalities may reflect interactions of high concentrations of adenosine with known adenosine A1 receptors in nervous tissue.5  A metabolic basis of CNS manifestations was strongly supported by findings in 2 clinical studies, where improvements of neurologic symptoms were noticed during enzyme replacement therapy using multiple partial-exchange transfusions, which resulted in a rapid reduction in concentrations of accumulated metabolites.5,33 

Although none of the patients in our study revealed remarkable neurologic abnormalities at diagnosis, our finding clearly indicates that there is a substantial risk of developing such abnormalities during further follow-up after HSCT, raising the important question as to why treatment by HSCT apparently fails to prevent this complication in a substantial proportion of patients. Similar to transfused red cells used in the studies mentioned, engrafted donor blood cells after transplantation are able to provide “enzyme replacement.” This was demonstrated in previous studies where marked improvement but no normalization of metabolic abnormalities was observed after HSCT.34,35  The variable neurologic outcome after HSCT cannot be explained by differences in the degree of metabolic correction as shown in this study. This perplexing variability is also demonstrated by our experience in the identical twins, where one is distinctly more severely affected neurologically than the other, although both received transplants simultaneously from the same donor. It should be pointed out that similar neurologic problems have so far not been reported in patients receiving long-term enzyme replacement with PEG-ADA. This therapy, which has been established since 1987 and has been used in more than 150 ADA-deficient patients, was shown to induce rapid and complete normalization of metabolic abnormalities.2,14,25  In fact, plasma ADA activity in patients on maintenance replacement therapy with PEG-ADA usually exceeds by several fold the total blood ADA activity in healthy individuals.25  The question thus must be raised whether this treatment approach may be more effective compared to HSCT with regard to protection of the CNS.

In summary, we report an unexpected high rate of neurologic complications in ADA-deficient patients after HSCT. We were unable to identify transplantation-related parameters correlating with this complication. It remains an important issue whether other treatment options such as regular enzyme replacement with PEG-ADA may offer advantages to prevent neurologic damage.

Contribution: M.H., M.-H.A., A.S., and W.F. designed and performed the study and prepared the initial draft of the manuscript; M.S.-S., C.S., B.B., T.G., and H.B. assisted in patient data collection; M.S.H., M.T.R., U.P., D.L., and K.S. provided laboratory data; all authors contributed to the discussion and the preparation of the final draft.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Wilhelm Friedrich, Universitätsklinik für Kinder- und Jugendmedizin, Eythstrasse 24, 89075 Ulm, Germany; e-mail: wilhelm.friedrich@uniklinik-ulm.de.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Tatjana Kersten and Gabi Keller for their technical assistance.

1
Buckley RH. Molecular defects in human severe combined immunodeficiency and approaches to immune reconstitution.
Annu Rev Immunol
2004
;
22
:
625
–655.
2
Hirschhorn R. Immunodeficiency disease due to deficiency of adenosine deaminase. In Ochs H (Ed.).
Primary immunodeficiency diseases: a molecular & cellular approach
1998
;Oxford, United Kingdom Oxford University Press pp.
121
–139.
3
Hershfield MS. New insights into adenosine-receptor-mediated immunosuppression and the role of adenosine in causing the immunodeficiency associated with adenosine deaminase deficiency.
Eur J Immunol
2005
;
35
:
25
–30.
4
Stephan JL, Vlekova V, Le Deist F, et al. Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients.
J Pediatr
1993
;
123
:
564
–572.
5
Hirschhorn R, Paageorgiou PS, Kesarwala HH, Taft LT. Amelioration of neurologic abnormalities after “enzyme replacement” in adenosine deaminase deficiency.
N Engl J Med
1980
;
303
:
377
–380.
6
Tanaka C, Hara T, Suzaki I, Maegaki Y, Takeshita K. Sensorineural deafness in siblings with adenosine deaminase deficiency.
Brain Dev
1996
;
18
:
304
–306.
7
Buckley RH, Schiff SE, Schiff RI, et al. Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency.
N Engl J Med
1999
;
340
:
508
–516.
8
Fischer A, Landais P, Friedrich W, et al. European experience of bone-marrow transplantation for severe combined immunodeficiency.
Lancet
1990
;
336
:
850
–854.
9
Dror Y, Gallagher R, Wara DW, et al. Immune reconstitution in severe combined immunodeficiency disease after lectin-treated, T-cell-depleted haplocompatible bone marrow transplantation.
Blood
1993
;
81
:
2021
–2030.
10
Friedrich W, Goldmann SF, Ebell W, et al. Severe combined immunodeficiency: treatment by bone marrow transplantation in 15 infants using HLA-haploidentical donors.
Eur J Pediatr
1985
;
144
:
125
–130.
11
O'Reilly RJ, Brochstein J, Collins N, et al. Evaluation of HLA-haplotype disparate parental marrow grafts depleted of T lymphocytes by differential agglutination with a soybean lectin and E-rosette depletion for the treatment of severe combined immunodeficiency.
Vox Sang
1986
;
51
:suppl 2,
81
–86.
12
Haddad E, Landais P, Friedrich W, et al. 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
1998
;
91
:
3646
–3653.
13
Hershfield MS. Adenosine deaminase deficiency: clinical expression, molecular basis, and therapy.
Semin Hematol
1998
;
35
:
291
–298.
14
Hershfield MS. PEG-ADA replacement therapy for adenosine deaminase deficiency: an update after 8.5 years.
Clin Immunol Immunopathol
1995
;
76
:
S228
–232.
15
Aiuti A, Ficara F, Cattaneo F, Bordignon C, Roncarolo MG. Gene therapy for adenosine deaminase deficiency.
Curr Opin Allergy Clin Immunol
2003
;
3
:
461
–466.
16
Aiuti A, Slavin S, Aker M, et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning.
Science
2002
;
296
:
2410
–2413.
17
Gaspar HB, Bjorkegren E, Parsley K, et al. Successful reconstitution of immunity in ADA-SCID by stem cell gene therapy following cessation of PEG-ADA and use of mild preconditioning.
Mol Ther
2006
;
14
:
505
–513.
18
Bluetters-Sawatzki R, Friedrich W, Ebell W, et al. HLA-haploidentical bone marrow transplantation in three infants with adenosine deaminase deficiency: stable immunological reconstitution and reversal of skeletal abnormalities.
Eur J Pediatr
1989
;
149
:
104
–109.
19
Hershfield MS, Fetter JE, Small WC, et al. Effects of mutational loss of adenosine kinase and deoxycytidine kinase on deoxyATP accumulation and deoxyadenosine toxicity in cultured CEM human T-lymphoblastoid cells.
J Biol Chem
1982
;
257
:
6380
–6386.
20
Hershfield MS, Kredich NM, Koller CA, et al. S-adenosylhomocysteine catabolism and basis for acquired resistance during treatment of T-cell acute lymphoblastic leukemia with 2′-deoxycoformycin alone and in combination with 9-beta-D-arabinofuranosyladenine.
Cancer Res
1983
;
43
:
3451
–3458.
21
Comans-Bitter WM, de Groot R, van den Beemd R, et al. Immunophenotyping of blood lymphocytes in childhood. Reference values for lymphocyte subpopulations.
J Pediatr
1997
;
130
:
388
–393.
22
Arredondo-Vega FX, Santisteban I, Daniels S, Toutain S, Hershfield MS. Adenosine deaminase deficiency: genotype-phenotype correlations based on expressed activity of 29 mutant alleles.
Am J Hum Genet
1998
;
63
:
1049
–1059.
23
Hirschhorn R, Beratis N, Rosen FS, Parkman R, Stern R, Polmar S. Adenosine-deaminase deficiency in a child diagnosed prenatally.
Lancet
1975
;
1
:
73
–75.
24
Valerio D, Dekker BM, Duyvesteyn MG, et al. One adenosine deaminase allele in a patient with severe combined immunodeficiency contains a point mutation abolishing enzyme activity.
EMBO J
1986
;
5
:
113
–119.
25
Hershfield M. Combined immune deficiencies due to purine enzyme defects. In Stiehm ER (Ed.).
Immunologic Disorders in Infants and Children
2004
; 5th ed Philadelphia, PA WB Saunders pp.
480
–504.
26
Hershfield MS. PEG-ADA: an alternative to haploidentical bone marrow transplantation and an adjunct to gene therapy for adenosine deaminase deficiency.
Hum Mutat
1995
;
5
:
107
–112.
27
Hershfield MS, Buckley RH, Greenberg ML, et al. Treatment of adenosine deaminase deficiency with polyethylene glycol-modified adenosine deaminase.
N Engl J Med
1987
;
316
:
589
–596.
28
Chan B, Wara D, Bastian J, et al. Long-term efficacy of enzyme replacement therapy for adenosine deaminase (ADA)-deficient severe combined Immunodeficiency (SCID).
Clin Immunol
2005
;
117
:
133
–143.
29
Malacarne F, Benicchi T, Notarangelo LD, et al. Reduced thymic output, increased spontaneous apoptosis and oligoclonal B cells in polyethylene glycol-adenosine deaminase-treated patients.
Eur J Immunol
2005
;
35
:
3376
–3386.
30
Small TN, Wall DA, Kurtzberg J, Cowan MJ, O'Reilly RJ, Friedrich W. Association of reticular dysgenesis (thymic alymphoplasia and congenital aleukocytosis) with bilateral sensorineural deafness.
J Pediatr
1999
;
135
:
387
–389.
31
Rogers MH, Lwin R, Fairbanks L, Gerritsen B, Gaspar HB. Cognitive and behavioral abnormalities in adenosine deaminase deficient severe combined immunodeficiency.
J Pediatr
2001
;
139
:
44
–50.
32
Keightley RG, Lawton AR, Cooper MD, Yunis EJ. Successful fetal liver transplantation in a child with severe combined immunodeficiency.
Lancet
1975
;
2
:
850
–853.
33
Polmar SH, Stern RC, Schwartz AL, Wetzler EM, Chase PA, Hirschhorn R. Enzyme replacement therapy for adenosine deaminase deficiency and severe combined immunodeficiency.
N Engl J Med
1976
;
295
:
1337
–1343.
34
Fairbanks LD, Simmonds HA, Duley JA, Gaspar HB, Flood T, Steward CA. ADA activity and DATP levels in erythrocytes after bone marrow transplantation.
Adv Exp Med Biol
2000
;
486
:
51
–55.
35
Hirschhorn R, Roegner-Maniscalco V, Kuritsky L, Rosen FS. Bone marrow transplantation only partially restores purine metabolites to normal in adenosine deaminase-deficient patients.
J Clin Invest
1981
;
68
:
1387
–1393.
Sign in via your Institution