Skip to main content
Erschienen in: Journal of Clinical Immunology 3/2011

Open Access 01.06.2011

Allogeneic Haematopoietic Stem Cell Transplantation as Therapy for Chronic Granulomatous Disease—Single Centre Experience

verfasst von: Jolanta Goździk, Anna Pituch-Noworolska, Szymon Skoczeń, Wojciech Czogała, Jarosław Baran, Aleksandra Krasowska-Kwiecień, Oktawiusz Wiecha, Marek Zembala

Erschienen in: Journal of Clinical Immunology | Ausgabe 3/2011

Abstract

Chronic granulomatous disease (CGD) is phagocytic cell metabolic disorder resulting in recurrent infections and granuloma formation. This paper reports the favourable outcome of allogeneic transplantation in six high-risk CGD patients. The following donors were used: HLA-matched, related (two) and unrelated (three), and HLA-mismatched, unrelated (one). One patient was transplanted twice using the same sibling donor because of graft rejection at 6 months after reduced-intensity conditioning transplant (fludarabine and melphalan). Myeloablative conditioning regimen consisted of busulphan and cyclophosphamide. Stem cell source was unmanipulated bone marrow containing: 5.2 (2.6–6.5) × 108 nucleated cells, 3.8 (2.0–8.0) × 106 CD34+ cells and 45 (27–64) × 106 CD3+ cells per kilogramme. Graft-versus-host disease prophylaxis consisted of cyclosporine A and, for unrelated donors, short course of methotrexate and anti-T-lymphocyte globulin. Mean neutrophile and platelet engraftments were observed at day 22 (20–23) and day 20 (16–29), respectively. Pre-existing infections and inflammatory granulomas resolved. With the follow-up of 4–35 months (mean, 20 months), all patients are alive and well with full donor chimerism and normalized superoxide production.
Hinweise
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1007/​s10875-011-9596-5

Introduction

Chronic granulomatous disease (CGD) is X-linked or autosomal recessive inherited immune deficiency caused by mutations in the genes encoding the various subunits of the NADPH oxidase system of phagocytic cells responsible for the killing of phagocytosed microorganisms. CGD occurs with an estimated frequency of one in 250,000 live births. This defect is associated with significant morbidity and mortality so the predicted life expectancy is of about 25–30 years of age. The disease is characterized by recurrent, severe and life-threatening, bacterial and fungal infections with granuloma formation in different organs [1].
Usual sites of infections, mainly caused by catalase-positive microorganisms (e.g. Aspergillus sp., Staphylococcus sp., Salmonella sp.), are lungs, skin, gastrointestinal tract, liver and lymph nodes. Persistent inflammatory reaction to infection may lead to colitis with subsequent growth failure, gastric outlet obstruction, lung disease and granuloma formation [2, 3].
Conventional management for patients with CGD consists of lifelong anti-infectious prophylaxis with cotrimoxazole or antibiotics with intracellular activity, antimycotics steroids and/or interferon-γ (IFN-γ). Nevertheless, the annual mortality is still 2–5%, and only 50% of patients are alive at 30 years [4, 5]. Therefore, there is a need for more effective therapies. Allogeneic haematopoietic stem cell transplantation (HSCT) is the only curative treatment for CGD with the excellent outcome noted in patients asymptomatic at transplantation. In a high-risk group of CGD patients (adults with organ dysfunction and/or patients with active inflammation and infections), the transplant-related mortality (TRM) about 30% is noted [6, 7].
Here, we report on six young CGD patients successfully treated with allogeneic HSCT with full correction of the phagocytic function before onset of organ dysfunction due to chronic inflammation.

Patients and Methods

Patient 1, a 2-year-old boy, was diagnosed at the age of 1.5 years to have X-linked CGD. He presented with recurrent lymphadenopathy, pneumonia, diarrhoea and skin infection. On admission, the generalized lymphadenopathy, mild hepatomegaly and multiple granulomas of the lungs were observed.
Patient 2, a 13-year-old girl, was diagnosed as having CGD at the age of 2 years. In the past, she had Salmonella Typhimurium gastroenteritis, recurrent lymphadenopathy, cervical abscess, sinusitis with Aspergillus fumigatus, frequent pneumonias, sepsis, herpes and varicella infections and multiple skin infections. She was admitted to our department before transplantation with sinusitis due to aspergillosis despite intensive antifungal treatment (amphotericin B and voriconasole).
Patient 3, a 5-year-old boy, was diagnosed with CGD at the age of 10 months after surgical removal of multiple abscesses of lymph nodes. His mother’s older brother and her first son died of severe infections in early childhood. He has been repeatedly hospitalized with skin abscesses, meningitis, gastroenteritis (Salmonella Typhimurium) and severe pneumonias.
Patient 4, a 3.5-year-old boy with X-linked CGD (Xp21.1, subtype X91+), was diagnosed 6 months ago. The family history indicated that his mother’s brother died at the age of 3 years because of infection. The patient experienced recurrent lung and skin infections. The CT abdominal scan performed at the age of 3 years due to fever and abdominal pain showed hepatomegaly with multiple nodular areas.
Patient 5, a 1.5-year-old boy, was diagnosed with X-linked CGD at the age of 6 months. He had in the past sinusitis, kidney, liver abscess and multiple skin infections.
Patient 6, a 2-yeard old boy with CGD, was diagnosed 6 months ago after serious gastroenterocolitis (Salmonella Typhimurium). This boy is a cousin of patient 3.

Diagnosis

In all patients, the diagnosis of CGD was established on the basis of clinical symptoms and the lack of chemiluminescence response to latex stimulation of blood measurement as described previously [8] and confirmed by nitroblue tetrazolium (NBT) reduction test [9].

Prophylaxis and Treatment

The antibacterial and antimycotic treatment or prophylaxis was introduced immediately after diagnosis of CGD and continued until transplantation.

Transplantation

The stem cell transplantation and collection of data before treatment were performed after written informed consent of parents and patient 2. The information on the beneficial effects of conventional antibacterial/antifungal prophylaxis/treatment, the risk of allografting, especially in the presence of overt infections or inflammatory symptoms, and a lack of sibling donors was also provided. Donor and recipient HLA matching was performed by molecular typing of HLA classes I (A, B, Cw) and II loci (DRB1, DQB1). Patients 1 and 6 underwent bone marrow transplantation from a sibling donor with HLA-identical genotype. Patients 2, 4 and 5 received HLA-identical bone marrow from unrelated donors, and patient 3 was transplanted with bone marrow from mismatched unrelated donor [patient’s HLA: A*0201*2402, B*3906*4402, Cw*0702*0704, DRB1*1601, DQ*0502; donor’s HLA: A*0201*2402, B*3901*4402, Cw*1203*0704, DRB1*1601, DQ*0502]. The characteristics of patients and donors are shown in Table I. Patients were nursed in a high-efficiency, particle-air-filtered protected environment. During transplantation period, leucocyte-depleted and irradiated blood products were used. Colistin at 100,000 U/kg/day was given as an oral gut decontamination. Prophylaxis regimen also included cotrimoxazole, acyclovir, fluconazole, heparin and, when needed, intravenous immunoglobulin (IVIG) substitution until immune reconstitution. Patient 2 with fungal sinusitis at transplantation was treated with voriconazole and caspofungin until engraftment. Five of six patients received busulphan-based myeloablative conditioning regimen, combined with cyclophosphamide. Patient 1 was conditioned according to reduced-intensity (RI) protocol with the use of fludarabine and melphalan. Details of conditioning regimens are shown in Table II. Stem cell source was unmanipulated bone marrow containing 4.6 (2.6–6.5) × 108 nucleated cells (NS), 3.3 (2.0–4.9) × 106 CD34+ cells and 44 (26–64) × 106 CD3+ cells per kilogramme of recipient body weight (b.w.). The cell doses for patient transplanted with an RI were as follows: 3.0 × 108 NS, 4.0 × 106 CD34+ and 26 × 106 CD3+ cells per kilogramme of body weight. Graft-versus-host disease (GvHD) prophylaxis consisted of cyclosporine A (CsA) beginning on day −1 (patients 1, 5 and 6) and on day −4 (patients 2, 3 and 4). The plasma level of CsA was maintained between 150 and 200 μg/mL, and this therapy was continued until 3 months (patients 1 and 6) and 6 months (patients 2, 3, 4 and 5). The CsA was tapered rapidly because of gradually decreasing chimerism in patient 1 and slowly in patients 2, 3, 4, 5 and 6. Additionally, patients transplanted from unrelated donors received short course of methotrexate at 10 mg/m2.
Table I
The characteristics of patients and donors
 
Type of donor
Age (years)
Gender
CMV status
EBV status
Blood groups AB0 (Rh)
R
D
R
D
R
D
R
D
R
D
Patient 1
MSD
2/3a
6/7a
M
F
+
+
+
+
A (−)
A (−)
Patient 2
MUD
13
26
F
F
+
+
+
0 (−)
0 (+)
Patient 3
MMUD
5
28
M
F
+
+
+
0 (+)
0 (+)
Patient 4
MUD
4
21
M
M
+
+
A (+)
A (−)
Patient 5
MUD
1
26
M
F
+
+
+
B (+)
0 (+)
Patient 6
MSD
2
12
M
F
+
+
+
0 (+)
B (+)
R recipient, D donor, CMV cytomegalovirus, EBV Epstein–Barr virus, MSD matched sibling donor, MUD matched unrelated donor, MMUD mismatched unrelated donor, + positive, negative
aAt second transplantation
Table II
Conditioning regimens
 
Regimen
Total dose
Daily dose
Administration
Days
Patient 1
Flu
150 mg/m2
30 mg/m2
i.v. in 30 min
−7, −6, −5, −4, −3
Mel
140 mg/m2
70 mg/m2
i.v. in 1 h
−3, −2
Patient 1a
Bu
20 mg/kg
5 mg/kg
p.o. q 6 h
−9, −8, −7, −6
Cy
200 mg/kg
50 mg/kg
i.v. in 1 h
−5, −4, −3, −2
ATGb
7.5 mg/kg
3.75 mg/kg
i.v. in 8–10 h
−3, −2
Patient 2
Bu
16 mg/kg
4 mg/kg
p.o. q 6 h
−9, −8, −7, −6
Cy
200 mg/kg
50 mg/kg
i.v. in 1 h
−5, −4, −3, −2
ATGb
10 mg/kg
2.5 mg/kg
i.v. in 8–10 h
−4, −3, −2, −1
Patient 3
Bu
20 mg/kg
5 mg/kg
p.o. q 6 h
−7, −6, −5, −4
Cy
120 mg/kg
60 mg/kg
i.v. in 1 h
−3, −2
ATGc
60 mg/kg
20 mg/kg
i.v. in 8–10 h
−3, −2, −1
Patient 4
Bu
20 mg/kg
5 mg/kg
p.o. q 6 h
−9, −8, −7, −6
Cy
200 mg/kg
50 mg/kg
i.v. in 1 h
−5, −4, −3, −2
ATGc
60 mg/kg
20 mg/kg
i.v. in 8–10 h
−3, −2, −1
Patient 5
Bu
20 mg/kg
5 mg/kg
p.o. q 6 h
−9, −8, −7, −6
Cy
200 mg/kg
50 mg/kg
i.v. in 1 h
−5, −4, −3, −2
ATGb
10 mg/kg
2.5 mg/kg
i.v. in 8–10 h
−3, −2, −1
Patient 6
Bu
20 mg/kg
5 mg/kg
p.o. q 6 h
−9, −8, −7, −6
Cy
200 mg/kg
50 mg/kg
i.v. in 1 h
−5, −4, −3, −2
Flu fludarabine, Mel melphalan, Bu busulphan, Cy cyclophosphamide, ATG rabbit anti-T-cell globulin
aAt second transplantation
bGenzyme
cFresenius
The chimerism was studied at +30, 100, 180 and 360 days after standard HSCT and every 1 month after non-myeloablative transplantation by karyotyping (fluorescence in situ hybridization) or analysis of informative microsatellite DNA sequences with the use of standard techniques. Neutrophil function and NK-, T- and B-cell reconstitution measurements were performed at the same time schedule and then every year. Lymphocyte subsets were measured by flow cytometry including CD3+, CD4+, CD8+, CD19+ and CD16+56+ cells. The presence of oxidase-positive neutrophils was detected by NBT tests. Immunoglobulin levels (IgG, IgA, IgM) were measured by nephelometry.

Results

Engraftment

Engraftment with full chimerism and functioning neutrophiles were observed in all patients. Haemopoietic recovery occurred within a median time of 22 days (range, 20–23 days) to neutrophile count >500/μL and within a median time of 20 days (range, 16–29 days) to platelet count >20,000/mL (Table III). The donor-derived haemopoiesis and the normal NBT reduction remained in five patients. In patient 1, after initial engraftment, chimerism fell to 14% despite cessation of CsA and donor lymphocyte infusion (DLI) in four increasing doses every 4–6 weeks: first, 0.2 × 106/kg; second, 1.0 × 106/kg; third, 1.8 × 106/kg and fourth, 5.0 × 106/kg. In this patient, after the first HSCT, the lymphadenopathy and granulomas resolved despite of slow rejection. The second transplant from the same sibling donor after myeloablative conditioning was performed 8 months after the first HSCT (Table II). Unmanipulated bone marrow containing, per kilogramme of recipient b.w., 6.0 × 108 NS, 8.0 × 106 CD34+ cells and 53 × 106 CD3+ cells was infused. Neutrophil engraftment occurred on day +23 and platelets on day +29 (Table III). Full donor chimerism and normal value of NBT test were observed on day +30.
Table III
Engraftment and chimerism
 
Engraftment (day after HSCT)
NBT
Donor’s chimerism
Neutrophil >500/μL
PLT >20/μL
+30 day (N, >0.1)
+30 days (%)
+100 days (%)
+180 days (%)
+1 year (%)
Patient 1
22
0.154
100
37
14
Patient 1a
23
29
0.226
100
100
100
100
Patient 2
20
17
0.318
100
100
100
100
Patient 3
21
21
0.207
100
100
100
100
Patient 4
22
18
0.218
100
100
100
100
Patient 5
22
16
0.315
100
100
Patient 6
21
29
0.143
100
100
HSCT haematopoietic stem cell transplantation, PLT platelets, NBT nitroblue tetrazolium test, N normal value
aAt second transplantation

Survival

There was no episode of serious conditioning-related toxicity. For patients 1, 2, 4, 5 and 6, transfusion requirement was low, with median of 2 (range, 1–3) red blood cell concentrates and median of 4 (range, 3–7) of platelet transfusions. Patient 3 received eight red blood cell and six platelet concentrates because of haemorrhagic diarrhoea due to acute GvHD (aGvHD). He had also rotavirus infection and cytomegalovirus reactivation treated with gancyclovir with good response. No febrile episodes or exacerbations of preexisting infections were observed. The median length of hospitalization was 36 days (range, 33–61 days). In patient 2, the short episode of haemorrhagic cystitis grade II was observed at 3 months after transplantation but resolved after symptomatic therapy. All patients are alive and well with a median follow-up of 20 months (range, 4–35 months) after HSCT. In all patients, the lymphadenopathy and granulomas resolved and therapy of refractory pre-existing infections is not required.

Graft-Versus-Host Disease

Two patients had symptoms of aGvHD. Patient 2 presented with mild aGvHD (increasing level of bilirubin and AAT) with good response to steroids. Patient 3 developed severe grade IV acute GvHD with the gut, liver and skin involvement that required therapy with prednisolone, mycophenolate mofetil, tacrolimus and oral budezonide, and which finally resolved after anti-TNF-α therapy (etanercept) and infusion of mesenchymal cells (0.3 × 106/kg). Again, the limited chronic GVHD of skin developed but gradually responded to treatment without continuing sequelae.

Immunological Reconstitution

Since engraftment, all patients demonstrated normal NBT test. The IVIG replacement therapy was discontinued shortly after transplantation. Patients transplanted from unrelated donors showed subnormal count of lymphocyte T, B and NK cells during first year after HSCT. The normalization of lymphocyte number was faster in patient transplanted from matched related donor. All patients were included in revaccination protocol according to our schedule [10, 11]. In patients 1 and 2, the basic programme of vaccination against tetanus, diphtheria and hepatitis B virus was completed. Antibody production against these pathogens was adequate.

Discussion

CGD has been extensively studied during the last years. Because gene therapy is still in its infancy, allogeneic HSCT remains the only curative therapy for CGD. The benefits of HSCT like normal growth and improvement in quality of life with no need for medication are clear. This is in contrast to non-transplanted patients who remain on lifelong antimicrobial prophylaxis, with a continued risk of infections resistant to the prophylactic treatment that require frequent hospitalization [5, 12].
Although the first reports of transplantation in CGD were not so optimistic, the use of matched sibling donor improved the survival (above 90%) especially in children without severe infections and at an early stage of disease. As the chance of finding a matched related donor is less than 25%, and haploidentical HSCT is considered as a high risk due to delayed immune reconstitution and graft failure, the unrelated donor transplantation has been established as an alternative procedure [13]. However, the use of unrelated donor could be associated with a higher risk of complications after HSCT [14].
In our study, four patients underwent transplantation from unrelated donors (three matched and one mismatched). This mismatched unrelated donor was used in situation of a serious clinical status of the recipient and after four years of unsuccessfully searching for a matched unrelated donor. Finally, the decision of mismatched donor HSCT was regarded as a life-saving procedure.
The EBMT advocated myeloablative regimens, mostly consisting of busulphan and cyclophosphamide allografts from HLA-matched related donors, which provided excellent results in low-risk CGD patients [15, 16]. In opinion of other researchers, the myeloablative conditioning was associated with the high rates of severe acute GVHD and pulmonary infections leading to the TRM of above 30% especially in advanced CGD patients with active inflammation or infections [6, 17].
The therapeutic option of allogeneic transplantation after reduced-intensity conditioning (RIC) may be the alternative for CGD patients with coexisting severe infections and organ damage [18, 19]. However, the RIC regimens performed until now have shown a significant risk of incomplete engraftment with the donor haematopoietic cells or graft rejection and GvHD, particularly if DLI has to be used to ensure engraftment. Nevertheless, the RIC HSCT is usually enough to improve clinical status and resolve the inflammation and infections before the graft rejection [2022]. Moreover, additional standard myeloablative HSCT could be performed as a salvage therapy if second transplant is required [23].
In the present group, patient 1 was transplanted twice from the HLA-identical sibling. The first time was with RIC and subsequently with myeloablative conditioning. After transplantation with RIC, the graft rejection occurred and donor’s chimerism decreased from 100% to 14% within 6 months despite withdrawal of immunosuppression and DLI. The subsequent transplantation led to a rapid engraftment. Five remaining patients who received myeloablative HSCT donors engrafted around 22 days after transplantation, and the stable full donor chimerism and normal phagocyte function were observed at 30 days.
In our opinion, for patients with CGD, the optimal time of transplantation is critical. In most cases of CGD, HSCT is postponed until the patient is chronically ill. However, if transplantation is delayed, the chances of severe infections, the risk of GVHD and other serious transplant complications significantly increase [6].
The severe GVHD remains a special risk in CGD patients, possibly because CGD phagocytes have a specific propensity for increased production of TNF-α. Moreover, its level significantly rose in patients with granulomatous colitis or aspergillosis. Therefore, TNF-antagonist therapy may be beneficial if given as early treatment for GVHD and other complications [24, 25].
All presented patients are now judged as cured by clinical status and phagocytic function. No conditioning toxicity was observed despite the use of myeloablative regimens.
We believe that it is desirable to perform HSCT in young patients with proven diagnosis of CGD before the onset of life-threatening infections and organ damage due to chronic inflammation. As the HSCT procedure is safe enough, it may challenge the common view that HSCT is indicated in CGD patients only after severe clinical episode confirming the diagnosis.

Acknowledgements

The work was supported in part by grants from the Committee of Scientific Research No. NN402108833.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Winkelstein JA, Marino MC, Johnston RB, Boyle J, Curnutte J, Gallin Jl, et al. Chronic granulomatous disease: report on a national registry of 368 patients. Medicine. 2000;79:155–69.PubMedCrossRef Winkelstein JA, Marino MC, Johnston RB, Boyle J, Curnutte J, Gallin Jl, et al. Chronic granulomatous disease: report on a national registry of 368 patients. Medicine. 2000;79:155–69.PubMedCrossRef
2.
Zurück zum Zitat Jones L, McGrogan P, Flood TJ, Gennery AR, Morton L, Thrasher A, et al. Chronic granulomatous disease in the UK and Ireland—a comprehensive national patient based registry. Clin Exp Immunol. 2008;152:211–18.PubMedCrossRef Jones L, McGrogan P, Flood TJ, Gennery AR, Morton L, Thrasher A, et al. Chronic granulomatous disease in the UK and Ireland—a comprehensive national patient based registry. Clin Exp Immunol. 2008;152:211–18.PubMedCrossRef
3.
Zurück zum Zitat Martire B, Rondelli R, Soresina A, Pignata C, Broccoletti T, Finocchi A, et al. Clinical features, long-term follow-up and outcome of a large cohort of patients with chronic granulomatous disease: an Italian multicenter study. Clin Immunol. 2008;126:155–64.PubMedCrossRef Martire B, Rondelli R, Soresina A, Pignata C, Broccoletti T, Finocchi A, et al. Clinical features, long-term follow-up and outcome of a large cohort of patients with chronic granulomatous disease: an Italian multicenter study. Clin Immunol. 2008;126:155–64.PubMedCrossRef
4.
Zurück zum Zitat Kobayashi S, Murayama S, Takanashi S, Takahashi K, Miyatsuka S, Fujita T, et al. Clinical features and prognoses of 23 patients with chronic granulomatous disease followed for 21 years by a single hospital in Japan. Eur J Pediatr. 2008;167:1389–94.PubMedCrossRef Kobayashi S, Murayama S, Takanashi S, Takahashi K, Miyatsuka S, Fujita T, et al. Clinical features and prognoses of 23 patients with chronic granulomatous disease followed for 21 years by a single hospital in Japan. Eur J Pediatr. 2008;167:1389–94.PubMedCrossRef
5.
Zurück zum Zitat Kang EM, Malech HL. Advances in treatment for chronic granulomatous disease. Immunol Res. 2009;43:77–84.PubMedCrossRef Kang EM, Malech HL. Advances in treatment for chronic granulomatous disease. Immunol Res. 2009;43:77–84.PubMedCrossRef
6.
Zurück zum Zitat Seger RA, Gungor T, Belohradsky BH, Blanche S, Bordigoni P, Di Bartolomeo P, et al. Treatment of chronic granulomatous disease with myeloablative conditioning and an unmodified hemopoietic allograft: a survey of the European experience, 1985–2000. Blood. 2002;100:4344–50.PubMedCrossRef Seger RA, Gungor T, Belohradsky BH, Blanche S, Bordigoni P, Di Bartolomeo P, et al. Treatment of chronic granulomatous disease with myeloablative conditioning and an unmodified hemopoietic allograft: a survey of the European experience, 1985–2000. Blood. 2002;100:4344–50.PubMedCrossRef
7.
Zurück zum Zitat Soncini E, Slatter MA, Jones LBKR, Hughes S, Hodges S, Flood TJ, et al. Unrelated donor and HLA-identical sibling haematopoietic stem cell transplantation cure chronic granulomatous disease with good long-term outcome and growth. Br J Haematol. 2009;145:73–83.PubMedCrossRef Soncini E, Slatter MA, Jones LBKR, Hughes S, Hodges S, Flood TJ, et al. Unrelated donor and HLA-identical sibling haematopoietic stem cell transplantation cure chronic granulomatous disease with good long-term outcome and growth. Br J Haematol. 2009;145:73–83.PubMedCrossRef
8.
Zurück zum Zitat Baran J, Weglarczyk K, Mysiak M, Guzik K, Ernst M, Flad HD, et al. Fas (CD95)-Fas ligand interactions are responsible for monocyte apoptosis occurring as a result of phagocytosis and killing of Staphylococcus aureus. Infect Immun. 2001;69:1287–97.PubMedCrossRef Baran J, Weglarczyk K, Mysiak M, Guzik K, Ernst M, Flad HD, et al. Fas (CD95)-Fas ligand interactions are responsible for monocyte apoptosis occurring as a result of phagocytosis and killing of Staphylococcus aureus. Infect Immun. 2001;69:1287–97.PubMedCrossRef
9.
Zurück zum Zitat Hellum KB. Nitroblue tetrazolium test in bacterial and viral infections. Scand J Infect Dis. 1977;9:269–76.PubMed Hellum KB. Nitroblue tetrazolium test in bacterial and viral infections. Scand J Infect Dis. 1977;9:269–76.PubMed
10.
Zurück zum Zitat Ljungman P, Engelhard D, de la Camara R, Einsele H, Locasciulli A, Martino R, et al. Vaccination of stem cell transplant recipients: recommendations of the infectious disease working party of the EBMT. Bone Marrow Transplant. 2005;35:737–46.PubMedCrossRef Ljungman P, Engelhard D, de la Camara R, Einsele H, Locasciulli A, Martino R, et al. Vaccination of stem cell transplant recipients: recommendations of the infectious disease working party of the EBMT. Bone Marrow Transplant. 2005;35:737–46.PubMedCrossRef
11.
Zurück zum Zitat Goździk J, Czajka H, Skoczeń Sz, Baran J, Czogała W, Tarczoń I, et al. Safety and efficacy revaccination of children after stem cell transplantation. Med Wieku Rozwoj. 2010;14:43–53. Goździk J, Czajka H, Skoczeń Sz, Baran J, Czogała W, Tarczoń I, et al. Safety and efficacy revaccination of children after stem cell transplantation. Med Wieku Rozwoj. 2010;14:43–53.
12.
Zurück zum Zitat Del Giudice I, Iori AP, Mengarelli A, Testi AM, Romano A, Cerretti R, et al. Allogeneic stem cell transplant from HLA-identical sibling for chronic granulomatous disease and review of the literature. Ann Hematol. 2003;82:189–92.PubMed Del Giudice I, Iori AP, Mengarelli A, Testi AM, Romano A, Cerretti R, et al. Allogeneic stem cell transplant from HLA-identical sibling for chronic granulomatous disease and review of the literature. Ann Hematol. 2003;82:189–92.PubMed
13.
Zurück zum Zitat Seger RA. Hematopoietic stem cell transplantation for chronic granulomatous disease. Immunol Allergy Clin North Am. 2010;30:195–208.PubMedCrossRef Seger RA. Hematopoietic stem cell transplantation for chronic granulomatous disease. Immunol Allergy Clin North Am. 2010;30:195–208.PubMedCrossRef
14.
Zurück zum Zitat Ottinger HD, Ferencik S, Beelen DW, Lindemann M, Peceny R, Elmaagacli AH, et al. Hematopoietic stem cell transplantation: contrasting the outcome of transplantations from HLA-identical siblings, partially HLA-mismatched related donors, and HLA-matched unrelated donors. Blood. 2003;102:1131–7.PubMedCrossRef Ottinger HD, Ferencik S, Beelen DW, Lindemann M, Peceny R, Elmaagacli AH, et al. Hematopoietic stem cell transplantation: contrasting the outcome of transplantations from HLA-identical siblings, partially HLA-mismatched related donors, and HLA-matched unrelated donors. Blood. 2003;102:1131–7.PubMedCrossRef
15.
Zurück zum Zitat Fisher A, Landais P, Friedrich W, Gerritsen B, Fasth A, Porta F, et al. Bone marrow transplantation (BMT) in Europe for primary immunodeficiencies other than severe combined immunodeficiency: a report from the European group for BMT and the European group for immunodeficiency. Blood. 1994;83:1149–54. Fisher A, Landais P, Friedrich W, Gerritsen B, Fasth A, Porta F, et al. Bone marrow transplantation (BMT) in Europe for primary immunodeficiencies other than severe combined immunodeficiency: a report from the European group for BMT and the European group for immunodeficiency. Blood. 1994;83:1149–54.
16.
Zurück zum Zitat Seger R, Flood T. Guidelines for BMT in chronic granulomatous disease (CGD). EBMT/ESID WP inborn errors: EBMT guidelines for haematopoietic stem cell transplantation for primary immunodeficiencies. Paris: EBMT; 2004. Seger R, Flood T. Guidelines for BMT in chronic granulomatous disease (CGD). EBMT/ESID WP inborn errors: EBMT guidelines for haematopoietic stem cell transplantation for primary immunodeficiencies. Paris: EBMT; 2004.
17.
Zurück zum Zitat Hara K, Kajiume T, Kondo T, Sera Y, Kawaguchi H, KobaYashi M. Respiratory complications after hematopoietic stem cell transplantation in patients with chronic granulomatous disease. Transfus Med. 2009;19:105–8.PubMedCrossRef Hara K, Kajiume T, Kondo T, Sera Y, Kawaguchi H, KobaYashi M. Respiratory complications after hematopoietic stem cell transplantation in patients with chronic granulomatous disease. Transfus Med. 2009;19:105–8.PubMedCrossRef
18.
Zurück zum Zitat Horwitz ME, Barrett AJ, Brown MR, Carter CS, Childs R, Gallin JI, et al. Treatment of chronic granulomatous disease with nonmyeloablative conditioning and a T-cell-depleted hematopoietic allograft. N Engl J Med. 2001;344:881–8.PubMedCrossRef Horwitz ME, Barrett AJ, Brown MR, Carter CS, Childs R, Gallin JI, et al. Treatment of chronic granulomatous disease with nonmyeloablative conditioning and a T-cell-depleted hematopoietic allograft. N Engl J Med. 2001;344:881–8.PubMedCrossRef
19.
Zurück zum Zitat Kikuta A, Ito M, Mochizuki K, Akaihata M, Nemoto K, Sano H, et al. Nonmyeloablative stem cell transplantation for nonmalignant disease in children with severe organ dysfunction. Bone Marrow Transplant. 2006;38:665–9.PubMedCrossRef Kikuta A, Ito M, Mochizuki K, Akaihata M, Nemoto K, Sano H, et al. Nonmyeloablative stem cell transplantation for nonmalignant disease in children with severe organ dysfunction. Bone Marrow Transplant. 2006;38:665–9.PubMedCrossRef
20.
Zurück zum Zitat Gungor T, Halter J, Klink A, Junge S. Successful low toxicity hematopoietic stem cell transplantation for high-risk adult chronic granulomatous disease patients. Transplantation. 2005;79:1596–606.PubMedCrossRef Gungor T, Halter J, Klink A, Junge S. Successful low toxicity hematopoietic stem cell transplantation for high-risk adult chronic granulomatous disease patients. Transplantation. 2005;79:1596–606.PubMedCrossRef
21.
Zurück zum Zitat Nagler A, Ackerstein A, Kapelushnik J, Or R, Naparstek E, Slavin S. Donor lymphocyte infusion post-non-myeloablative allogeneic peripheral blood stem cell transplantation for chronic granulomatous disease. Bone Marrow Transplant. 1999;24:339–42.PubMedCrossRef Nagler A, Ackerstein A, Kapelushnik J, Or R, Naparstek E, Slavin S. Donor lymphocyte infusion post-non-myeloablative allogeneic peripheral blood stem cell transplantation for chronic granulomatous disease. Bone Marrow Transplant. 1999;24:339–42.PubMedCrossRef
22.
Zurück zum Zitat Schuetz C, Hoenig M, Schulz A, Lee-Kirsch MA, Roesler J, Friedrich W, et al. Successful unrelated bone marrow transplantation in a child with chronic granulomatous disease complicated by pulmonary and cerebral granuloma formation. Eur J Pediatr. 2007;166:785–8.PubMedCrossRef Schuetz C, Hoenig M, Schulz A, Lee-Kirsch MA, Roesler J, Friedrich W, et al. Successful unrelated bone marrow transplantation in a child with chronic granulomatous disease complicated by pulmonary and cerebral granuloma formation. Eur J Pediatr. 2007;166:785–8.PubMedCrossRef
23.
Zurück zum Zitat Nicholson JAT, Wynn RF, Carr TF, Will AM. Sequential reduced- and full-intensity allografting using same donor in a child with chronic granulomatous disease and coexistent, significant comorbidity. Bone Marrow Transplant. 2004;34:1009–10.PubMedCrossRef Nicholson JAT, Wynn RF, Carr TF, Will AM. Sequential reduced- and full-intensity allografting using same donor in a child with chronic granulomatous disease and coexistent, significant comorbidity. Bone Marrow Transplant. 2004;34:1009–10.PubMedCrossRef
24.
Zurück zum Zitat Morgenstern DE, Gifford MAC, LinLi L, Doerschuk CM, Dinauer MC. Absence of respiratory burst in X-linked chronic granulomatous disease and inflammatory response to Aspergillus fumigatus. J Exp Med. 1997;185:207–18.PubMedCrossRef Morgenstern DE, Gifford MAC, LinLi L, Doerschuk CM, Dinauer MC. Absence of respiratory burst in X-linked chronic granulomatous disease and inflammatory response to Aspergillus fumigatus. J Exp Med. 1997;185:207–18.PubMedCrossRef
25.
Zurück zum Zitat Hill GR, Teshima T, Rebel VI, Krijanowski Ol, Cookie KR, Brinson YS, et al. The p55 TNF-alpha receptor plays a critical role in T cell alloreactivity. J Immunol. 2000;164:656–63.PubMed Hill GR, Teshima T, Rebel VI, Krijanowski Ol, Cookie KR, Brinson YS, et al. The p55 TNF-alpha receptor plays a critical role in T cell alloreactivity. J Immunol. 2000;164:656–63.PubMed
Metadaten
Titel
Allogeneic Haematopoietic Stem Cell Transplantation as Therapy for Chronic Granulomatous Disease—Single Centre Experience
verfasst von
Jolanta Goździk
Anna Pituch-Noworolska
Szymon Skoczeń
Wojciech Czogała
Jarosław Baran
Aleksandra Krasowska-Kwiecień
Oktawiusz Wiecha
Marek Zembala
Publikationsdatum
01.06.2011
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 3/2011
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-011-9513-y

Weitere Artikel der Ausgabe 3/2011

Journal of Clinical Immunology 3/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.