Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Sci Immunol ; 5(44)2020 02 28.
Article in English | MEDLINE | ID: mdl-32111619

ABSTRACT

We investigated the molecular and cellular basis of severe combined immunodeficiency (SCID) in six patients with otofaciocervical syndrome type 2 who failed to attain T cell reconstitution after allogeneic hematopoietic stem cell transplantation, despite successful engraftment in three of them. We identified rare biallelic PAX1 rare variants in all patients. We demonstrated that these mutant PAX1 proteins have an altered conformation and flexibility of the paired box domain and reduced transcriptional activity. We generated patient-derived induced pluripotent stem cells and differentiated them into thymic epithelial progenitor cells and found that they have an altered transcriptional profile, including for genes involved in the development of the thymus and other tissues derived from pharyngeal pouches. These results identify biallelic, loss-of-function PAX1 mutations as the cause of a syndromic form of SCID due to altered thymus development.


Subject(s)
Paired Box Transcription Factors/immunology , Thymus Gland/immunology , Branchio-Oto-Renal Syndrome/genetics , Branchio-Oto-Renal Syndrome/immunology , Branchio-Oto-Renal Syndrome/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Infant , Male , Paired Box Transcription Factors/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , Thymus Gland/pathology
2.
Blood ; 130(25): 2728-2738, 2017 12 21.
Article in English | MEDLINE | ID: mdl-28935695

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome comprising familial/genetic HLH (FHL) and secondary HLH. In the HLH-94 study, with an estimated 5-year probability of survival (pSu) of 54% (95% confidence interval, 48%-60%), systemic therapy included etoposide, dexamethasone, and, from week 9, cyclosporine A (CSA). Hematopoietic stem cell transplantation (HSCT) was indicated in patients with familial/genetic, relapsing, or severe/persistent disease. In HLH-2004, CSA was instead administered upfront, aiming to reduce pre-HSCT mortality and morbidity. From 2004 to 2011, 369 children aged <18 years fulfilled HLH-2004 inclusion criteria (5 of 8 diagnostic criteria, affected siblings, and/or molecular diagnosis in FHL-causative genes). At median follow-up of 5.2 years, 230 of 369 patients (62%) were alive (5-year pSu, 61%; 56%-67%). Five-year pSu in children with (n = 168) and without (n = 201) family history/genetically verified FHL was 59% (52%-67%) and 64% (57%-71%), respectively (familial occurrence [n = 47], 58% [45%-75%]). Comparing with historical data (HLH-94), using HLH-94 inclusion criteria, pre-HSCT mortality was nonsignificantly reduced from 27% to 19% (P = .064 adjusted for age and sex). Time from start of therapy to HSCT was shorter compared with HLH-94 (P =020 adjusted for age and sex) and reported neurological alterations at HSCT were 22% in HLH-94 and 17% in HLH-2004 (using HLH-94 inclusion criteria). Five-year pSu post-HSCT overall was 66% (verified FHL, 70% [63%-78%]). Additional analyses provided specific suggestions on potential pre-HSCT treatment improvements. HLH-2004 confirms that a majority of patients may be rescued by the etoposide/dexamethasone combination but intensification with CSA upfront, adding corticosteroids to intrathecal therapy, and reduced time to HSCT did not improve outcome significantly.


Subject(s)
Dexamethasone/therapeutic use , Etoposide/therapeutic use , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/mortality , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclosporine/therapeutic use , Female , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Longitudinal Studies , Male , Treatment Outcome
4.
Biol Blood Marrow Transplant ; 22(4): 690-697, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26740373

ABSTRACT

The biology of engraftment syndrome is poorly understood, and the degree of overlap with acute graft-versus-host disease (GVHD) is unclear. To understand engraftment syndrome better, plasma cytokine profiles were evaluated in 56 pediatric allogeneic bone marrow transplant recipients before transplant, on the day of stem cell infusion, and weekly until day +100. Patients were divided into 4 groups: those with isolated engraftment syndrome (n = 8), acute GVHD (n = 12), both engraftment syndrome and acute GVHD (n = 4), and neither engraftment syndrome nor acute GVHD (n = 32). Engraftment syndrome was observed a median of 13.5 days (range, 10 to 28) after transplant, whereas acute GVHD was diagnosed a median of 55 days (range, 19 to 95) after transplant. Four patients developed both engraftment syndrome at a median of 10.5 days (range, 10 to 11) and acute GVHD at a median of 35 days (range, 23 to 56) after stem cell infusion. Median plasma levels of IL-1ß, IL-6, IL-12, IL-4, and IL-13 were significantly elevated in patients with isolated engraftment syndrome when compared with isolated acute GVHD. A rise of proinflammatory cytokines (IL-1ß, IL-6, and IL-12) was followed by surge in anti-inflammatory cytokines (IL-4 and IL-13) in patients with isolated engraftment syndrome. The observation of elevated IL-1ß suggests that engraftment syndrome could be an inflammasome mediated phenomenon.


Subject(s)
Cytokines/biosynthesis , Graft vs Host Disease/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Transplantation Conditioning , Acute Disease , Adolescent , Alemtuzumab , Antibodies, Monoclonal, Humanized/therapeutic use , Antilymphocyte Serum/therapeutic use , Child , Child, Preschool , Cyclosporine/therapeutic use , Cytokines/immunology , Female , Graft Survival , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Humans , Infant , Inflammasomes/immunology , Male , Myeloablative Agonists/therapeutic use , Prospective Studies , Survival Analysis , Transplantation, Homologous , Young Adult
7.
Blood ; 127(4): 503-12, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26644451

ABSTRACT

Reduced intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (HCT) with alemtuzumab, fludarabine, and melphalan is an effective approach for patients with nonmalignant disorders. Mixed chimerism and graft-versus-host-disease (GVHD) remain limitations on success. We hypothesized that higher levels of alemtuzumab at day 0 would result in a low risk of acute GVHD, a higher risk of mixed chimerism, and delayed early lymphocyte recovery and that alemtuzumab level thresholds for increased risks of these outcomes would be definable. We collected data from 105 patients to examine the influence of peritransplant alemtuzumab levels on acute GVHD, mixed chimerism, and lymphocyte recovery. The cumulative incidences of initial grades I-IV, II-IV, and III-IV acute GVHD in patients with alemtuzumab levels ≤0.15 vs ≥0.16 µg/mL were 68% vs 18% (P < .0001), 47% vs 13% (P = .0002), and 32% vs 8%, respectively (P = .005). The cumulative incidence of mixed chimerism in patients with an alemtuzumab level ≤0.15 µg/mL was 21%, vs 42% with levels of 0.16 to 4.35 µg/mL, and 100% with levels >4.35 µg/mL (P = .003). Patients with alemtuzumab levels ≤0.15 or 0.16 to 0.56 µg/mL had higher lymphocyte counts at day +30 and higher T-cell counts at day +100 compared with patients with levels ≥0.57 µg/mL (all P < .05). We conclude that peritransplant alemtuzumab levels impact acute GVHD, mixed chimerism, and lymphocyte recovery following RIC HCT with alemtuzumab, fludarabine, and melphalan. Precision dosing trials are warranted. We recommend a day 0 therapeutic range of 0.2 to 0.4 µg/mL.


Subject(s)
Antibodies, Monoclonal, Humanized/blood , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Melphalan/blood , Transplantation Conditioning , Vidarabine/analogs & derivatives , Adolescent , Adult , Alemtuzumab , Antibodies, Monoclonal, Humanized/therapeutic use , Child , Child, Preschool , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Lymphocytes/drug effects , Melphalan/therapeutic use , Prospective Studies , Transplantation Chimera , Transplantation Conditioning/adverse effects , Vidarabine/blood , Vidarabine/therapeutic use , Young Adult
8.
Hematol Oncol Clin North Am ; 29(5): 895-902, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26461149

ABSTRACT

Hemophagocytic Lymphohistiocytosis (HLH), an inherited life-threatening inflammatory disorder, has gained growing recognition not only in children but also increasingly in adults over the past 2 decades. HLH involves inborn defects in lymphocytes, which normally mediate control of infectious and inflammatory conditions within the immune system and in other tissues. In the context of inherited defects in cytotoxic cells and other immune cells, the disorder is classified as familial or primary HLH. Secondary HLH occurs in the settings of infections or underlying rheumatologic disorders. Secondary HLH also accompanies some lymphoid malignancies.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/etiology , Combined Modality Therapy , Genetic Predisposition to Disease , Genetic Variation , Humans , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/mortality , Lymphohistiocytosis, Hemophagocytic/therapy , Signal Transduction , Treatment Outcome
11.
Science ; 349(6246): 436-40, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26206937

ABSTRACT

Mutations in the LRBA gene (encoding the lipopolysaccharide-responsive and beige-like anchor protein) cause a syndrome of autoimmunity, lymphoproliferation, and humoral immune deficiency. The biological role of LRBA in immunologic disease is unknown. We found that patients with LRBA deficiency manifested a dramatic and sustained improvement in response to abatacept, a CTLA4 (cytotoxic T lymphocyte antigen-4)-immunoglobulin fusion drug. Clinical responses and homology of LRBA to proteins controlling intracellular trafficking led us to hypothesize that it regulates CTLA4, a potent inhibitory immune receptor. We found that LRBA colocalized with CTLA4 in endosomal vesicles and that LRBA deficiency or knockdown increased CTLA4 turnover, which resulted in reduced levels of CTLA4 protein in FoxP3(+) regulatory and activated conventional T cells. In LRBA-deficient cells, inhibition of lysosome degradation with chloroquine prevented CTLA4 loss. These findings elucidate a mechanism for CTLA4 trafficking and control of immune responses and suggest therapies for diseases involving the CTLA4 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autoimmune Diseases/drug therapy , CTLA-4 Antigen/deficiency , Common Variable Immunodeficiency/drug therapy , Immunoconjugates/therapeutic use , Abatacept , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Autoimmune Diseases/metabolism , CTLA-4 Antigen/genetics , Child , Chloroquine/pharmacology , Common Variable Immunodeficiency/metabolism , Endosomes/metabolism , Female , Forkhead Transcription Factors/analysis , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/metabolism , Lymphocyte Activation , Lysosomes/metabolism , Male , Proteolysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Young Adult
12.
Biol Blood Marrow Transplant ; 21(8): 1460-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25865646

ABSTRACT

Alemtuzumab, fludarabine, and melphalan reduced-intensity conditioning (RIC) regimens are increasingly used for the hematopoietic cell transplantation (HCT) of pediatric and young adult patients with nonmalignant diseases. Early experience suggests that these regimens are associated with good survival but a high incidence of mixed chimerism, which we have previously shown to be influenced by the alemtuzumab schedule. We hypothesized that the underlying diagnosis and donor graft source would also affect the development of mixed chimerism and that the majority of patients would survive RIC HCT without graft loss. To examine this, we conducted a retrospective study of 206 patients with metabolic diseases, non-Fanconi anemia marrow failure disorders, and primary immune deficiencies who underwent 210 consecutive RIC HCT procedures at Cincinnati Children's Hospital. Ninety-seven percent of the patients engrafted. Mixed donor and recipient chimerism developed in 46% of patients. Patients with marrow failure had a low risk of mixed chimerism (hazard ratio [HR], .208; 95% confidence interval [CI], .061 to .709; P = .012). The risk of mixed chimerism was high in patients who received a cord blood graft (HR, 3.122; 95% CI, 1.236 to 7.888; P = .016). As expected, patients who received a proximal or higher dose per kilogram of alemtuzumab schedule also experienced higher rates of mixed chimerism (all HR > 2, all P < .05). At the time of last follow-up (median, 654 days; range, 13 to 3337), over 75% of patients had greater than 90% whole blood donor chimerism. A second transplantation was performed in 5% of patients. Three-year survival without retransplantation was 84% (95% CI, 71% to 98%) for patients who underwent transplantation with an HLA-matched sibling donor. Survival without retransplantation was negatively affected by lack of a matched related donor, increasing age, and development of grades III and IV acute graft-versus-host disease. We conclude that alemtuzumab, fludarabine, and melphalan RIC HCT offers good results for many patients and that the risk of developing mixed chimerism is influenced by underlying diagnosis, graft source, and alemtuzumab dosing.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melphalan/therapeutic use , Vidarabine/analogs & derivatives , Adolescent , Adult , Alemtuzumab , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Child , Child, Preschool , Chimerism , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Melphalan/administration & dosage , Transplantation Conditioning/methods , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/therapeutic use , Young Adult
13.
Biol Blood Marrow Transplant ; 21(7): 1215-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25881755

ABSTRACT

Acute graft-versus-host disease (aGVHD) is mediated by allogeneic T cell responses. We hypothesized that increases of peripheral blood-activated CD8+ effector memory T (TEM) cells would be observed after hematopoietic stem cell transplantation (HSCT) before onset of aGVHD symptoms. Blood was collected twice weekly after HSCT for 7 weeks in 49 consecutive pediatric and adult HSCT recipients. Samples were incubated with fluorochrome-conjugated antibodies against CD45, CD3, CD8, CD38, CD45RA, and CCR7 and analyzed using flow cytometry. TEM cells were defined as CD3+ CD8+ CCR7- CD45RA(-) lymphocytes. CD38 expression was used as a marker of T cell activation. Patients were followed for 100 days for development of aGVHD. Twenty-three patients developed grade 1 to 4 aGVHD at a median of 37 days (range, 15 to 79 days) after HCST. Absolute CD38 bright CD8+ TEM of > 35 cells/µL predicted aGVHD at a median of 8 days (range, 1 to 34) before aGVHD onset with a sensitivity of 82.6% and specificity of 91.6%. The cumulative incidence of aGVHD was 90% in patients with absolute CD38 bright CD8+ TEM >35 cells/µL and 15% in patients without (P < .0001). Quantification of CD38 bright CD8+ TEM cells may predict aGVHD in children and young adult HSCT recipients.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , CD8-Positive T-Lymphocytes/immunology , Common Variable Immunodeficiency/therapy , Graft vs Host Disease/diagnosis , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Membrane Glycoproteins/immunology , Transplantation Conditioning , ADP-ribosyl Cyclase 1/genetics , Acute Disease , Adolescent , Adult , Biomarkers/analysis , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/pathology , Female , Gene Expression , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Histocompatibility Testing , Humans , Immunologic Memory , Immunophenotyping , Immunosuppressive Agents/therapeutic use , Infant , Lymphocyte Activation , Male , Membrane Glycoproteins/genetics , Myeloablative Agonists/therapeutic use , Transplantation, Homologous
14.
Br J Haematol ; 169(5): 711-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25817915

ABSTRACT

Patients with Langerhans cell histiocytosis (LCH) refractory to conventional chemotherapy have a poor outcome. There are currently two promising treatment strategies for high-risk patients: the first involves the combination of 2-chlorodeoxyadenosine and cytarabine; the other approach is allogeneic haematopoietic stem cell transplantation (HSCT). Here we evaluated 87 patients with high-risk LCH who were transplanted between 1990 and 2013. Prior to the year 2000, most patients underwent HSCT following myeloablative conditioning (MAC): only 5 of 20 patients (25%) survived with a high rate (55%) of transplant-related mortality (TRM). After the year 2000 an increasing number of patients underwent HSCT with reduced intensity conditioning (RIC): 49/67 (73%) patients survived, however, the improved survival was not overtly achieved by the introduction of RIC regimens with similar 3-year probability of survival after MAC (77%) and RIC transplantation (71%). There was no significant difference in TRM by conditioning regimen intensity but relapse rates were higher after RIC compared to MAC regimens (28% vs. 8%, P = 0·02), although most patients relapsing after RIC transplantation could be salvaged with further chemotherapy. HSCT may be a curative approach in 3 out of 4 patients with high risk LCH refractory to chemotherapy: the optimal choice of HSCT conditioning remains uncertain.


Subject(s)
Hematopoietic Stem Cell Transplantation , Histiocytosis, Langerhans-Cell/therapy , Transplantation Conditioning , Adolescent , Adult , Child , Child, Preschool , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Histiocytosis, Langerhans-Cell/mortality , Histiocytosis, Langerhans-Cell/pathology , Humans , Infant , Male , Retrospective Studies , Transplantation, Homologous , Treatment Outcome , Young Adult
15.
Biol Blood Marrow Transplant ; 21(2): 288-92, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25464116

ABSTRACT

Mixed donor chimerism is increasingly common in the pediatric hematopoietic stem cell transplantation (HSCT) setting because of the increased use of reduced-intensity preparative regimens for nonmalignant diseases. Donor lymphocyte infusion (DLI) is potentially useful in the treatment of mixed donor chimerism, but little are data available on the use of DLI in this setting. We conducted a retrospective review of 27 pediatric patients who received DLI for mixed donor chimerism between January 2006 and December 2010 after receiving a preparative regimen of alemtuzumab, fludarabine, and melphalan. Twenty-one patients (78%) were alive at a median of 35 months post-transplant. Seven patients (26%) sustained full donor chimerism after DLI only at a median of 35 months post-HSCT. Nine patients (33%) continued with mixed donor chimerism (median, 38% [range, 18% to 70%]) at a median of 37 months after DLI only. Five patients underwent unconditioned stem cell boosts or second conditioned transplants after no improvement in donor chimerism was seen following DLI. Donor source appeared to contribute to outcomes after DLI; patients with mismatched unrelated donors had earlier first decline in chimerism and timing of first DLI, a higher response rate to DLI, and an increased rate of graft-versus-host disease (GVHD). There was no response to DLI in patients with matched sibling donors. Ten patients, all with improvement in chimerism after DLI, developed acute GVHD after DLI, with 3 having grade III GVHD. Three patients developed chronic GVHD after DLI. These data illustrate the potential efficacy of DLI in the treatment of mixed donor chimerism after a reduced-intensity preparative regimen.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lymphocyte Transfusion , Lymphohistiocytosis, Hemophagocytic/therapy , Lymphoproliferative Disorders/therapy , Mucopolysaccharidoses/therapy , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning/methods , Adolescent , Alemtuzumab , Antibodies, Monoclonal, Humanized/therapeutic use , Child , Child, Preschool , Chimerism , Female , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/pathology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Melphalan/therapeutic use , Mucopolysaccharidoses/immunology , Mucopolysaccharidoses/pathology , Myeloablative Agonists/therapeutic use , Retrospective Studies , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , Siblings , Transplantation, Homologous , Treatment Outcome , Unrelated Donors , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
16.
J Clin Immunol ; 35(1): 15-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25388447

ABSTRACT

X-linked thrombocytopenia (XLT) is a mild form of the Wiskott-Aldrich syndrome (WAS) caused by mutations in the WAS gene. A recent retrospective study of the clinical outcome and molecular basis of a large cohort of XLT patients demonstrated that although overall survival is excellent, event free survival is severely affected with conservative treatment. To answer the question whether hematopoietic stem cell transplantation (HSCT) offers a viable alternative therapeutic option in XLT, we retrospectively investigated the outcome of HSCT in a cohort of 24 XLT patients who received HSCT between 1990 and 2011 at 14 transplant centers in the United States, Italy, Germany, Canada, and Japan. The engraftment rate was 100% and the overall survival rate was 83.3%. Of the four non-survivors, 2 underwent splenectomy prior to HSCT and died of sepsis, and two of aspergillus infections associated with severe GVHD. In all but one patient, pretransplant complications were resolved by HSCT. Our data indicate that HSCT following myeloablative conditioning is curative and associated with acceptable risks as a treatment option for XLT.


Subject(s)
Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Hematopoietic Stem Cell Transplantation , Mutation , Thrombocytopenia/genetics , Thrombocytopenia/therapy , Wiskott-Aldrich Syndrome Protein/genetics , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Disease-Free Survival , Genetic Diseases, X-Linked/mortality , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Kaplan-Meier Estimate , Male , Retrospective Studies , Thrombocytopenia/mortality , Wiskott-Aldrich Syndrome/genetics , Young Adult
17.
N Engl J Med ; 371(15): 1407-17, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25295500

ABSTRACT

BACKGROUND: In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS: We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS: All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS: This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Subject(s)
Gammaretrovirus/genetics , Genetic Therapy , Genetic Vectors , X-Linked Combined Immunodeficiency Diseases/therapy , Animals , Antigens, CD34 , DNA, Complementary/therapeutic use , Gene Expression , Gene Silencing , Genetic Therapy/adverse effects , Humans , Infant , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mutation , T-Lymphocytes/immunology , Transduction, Genetic , Transgenes/physiology , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
18.
N Engl J Med ; 371(5): 434-46, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25075835

ABSTRACT

BACKGROUND: The Primary Immune Deficiency Treatment Consortium was formed to analyze the results of hematopoietic-cell transplantation in children with severe combined immunodeficiency (SCID) and other primary immunodeficiencies. Factors associated with a good transplantation outcome need to be identified in order to design safer and more effective curative therapy, particularly for children with SCID diagnosed at birth. METHODS: We collected data retrospectively from 240 infants with SCID who had received transplants at 25 centers during a 10-year period (2000 through 2009). RESULTS: Survival at 5 years, freedom from immunoglobulin substitution, and CD3+ T-cell and IgA recovery were more likely among recipients of grafts from matched sibling donors than among recipients of grafts from alternative donors. However, the survival rate was high regardless of donor type among infants who received transplants at 3.5 months of age or younger (94%) and among older infants without prior infection (90%) or with infection that had resolved (82%). Among actively infected infants without a matched sibling donor, survival was best among recipients of haploidentical T-cell-depleted transplants in the absence of any pretransplantation conditioning. Among survivors, reduced-intensity or myeloablative pretransplantation conditioning was associated with an increased likelihood of a CD3+ T-cell count of more than 1000 per cubic millimeter, freedom from immunoglobulin substitution, and IgA recovery but did not significantly affect CD4+ T-cell recovery or recovery of phytohemagglutinin-induced T-cell proliferation. The genetic subtype of SCID affected the quality of CD3+ T-cell recovery but not survival. CONCLUSIONS: Transplants from donors other than matched siblings were associated with excellent survival among infants with SCID identified before the onset of infection. All available graft sources are expected to lead to excellent survival among asymptomatic infants. (Funded by the National Institute of Allergy and Infectious Diseases and others.).


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency/therapy , CD3 Complex/blood , Female , Graft vs Host Disease/epidemiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Immunoglobulin A/blood , Incidence , Infant , Lymphocyte Count , Male , Retreatment , Retrospective Studies , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/mortality , Siblings , Survival Rate , T-Lymphocytes/immunology , Transplantation Conditioning , Treatment Outcome
19.
Biol Blood Marrow Transplant ; 20(10): 1654-9, 2014 10.
Article in English | MEDLINE | ID: mdl-24979732

ABSTRACT

Therapy of refractory autoimmunity remains challenging. In this study, we evaluated the therapeutic effect of bortezomib, a proteasome inhibitor, by targeting plasma cells in 7 patients (median age, 9.9 years). Four doses of bortezomib were administered at a dose of 1.3 mg/m(2) intravenously (n = 6) or subcutaneously (n = 1) every 72 hours. Bortezomib was administered at a median of 120 days from laboratory confirmation of autoantibodies. All patients had failed 2 or more standard therapies. Rituximab was administered on the first day if B cells were present, and all patients received plasmapheresis 2 hours before bortezomib administration. Six patients experienced resolution of cytopenias. Two of 6 patients experienced recurrence of cytopenias after initial response. Adverse effects include nausea (n = 1), thrombocytopenia (n = 2), Clostridium difficile colitis (n = 1)), febrile neutropenia (n = 1), and cellulitis at the subcutaneous injection site (n = 1). Our experience suggests that bortezomib may be beneficial in the treatment of refractory autoimmunity in children.


Subject(s)
Anemia, Hemolytic, Autoimmune/drug therapy , Autoimmunity/drug effects , Boronic Acids/therapeutic use , Neutropenia/drug therapy , Proteasome Inhibitors/therapeutic use , Pyrazines/therapeutic use , Thrombocytopenia/drug therapy , Adolescent , Adult , Anemia, Hemolytic, Autoimmune/immunology , Anemia, Hemolytic, Autoimmune/pathology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Agents/therapeutic use , Bortezomib , Child , Child, Preschool , Drug Administration Schedule , Female , Humans , Infant , Male , Neutropenia/immunology , Neutropenia/pathology , Plasma Cells/drug effects , Plasma Cells/immunology , Plasma Cells/pathology , Plasmapheresis , Retrospective Studies , Rituximab , Thrombocytopenia/immunology , Thrombocytopenia/pathology
20.
Arthritis Rheumatol ; 66(12): 3486-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25047945

ABSTRACT

OBJECTIVE: Macrophage activation syndrome (MAS), a life-threatening complication of systemic juvenile idiopathic arthritis (JIA), resembles familial hemophagocytic lymphohistiocytosis (HLH), a constellation of autosomal-recessive immune disorders resulting from deficiency in cytolytic pathway proteins. We undertook this study to test our hypothesis that MAS predisposition in systemic JIA could be attributed to rare gene sequence variants affecting the cytotolytic pathway. METHODS: Whole-exome sequencing was used in 14 patients with systemic JIA and MAS and in their parents to identify protein-altering single-nucleotide polymorphisms/indels in known HLH-associated genes. To discover new candidate genes, the entire whole-exome sequencing data were filtered to identify protein-altering, rare recessive homozygous, compound heterozygous, and de novo variants with the potential to affect the cytolytic pathway. RESULTS: Heterozygous protein-altering rare variants in the known genes (LYST,MUNC13-4, and STXBP2) were found in 5 of 14 patients with systemic JIA and MAS (35.7%). This was in contrast to only 4 variants in 4 of 29 patients with systemic JIA without MAS (13.8%). Homozygosity and compound heterozygosity analysis applied to the entire whole-exome sequencing data in systemic JIA/MAS revealed 3 recessive pairs in 3 genes and compound heterozygotes in 73 genes. We also identified 20 heterozygous rare protein-altering variants that occurred in at least 2 patients. Many of the identified genes encoded proteins with a role in actin and microtubule reorganization and vesicle-mediated transport. "Cellular assembly and organization" was the top cellular function category based on Ingenuity Pathways Analysis (P < 3.10 × 10(-5) ). CONCLUSION: Whole-exome sequencing performed in patients with systemic JIA and MAS identified rare protein-altering variants in known HLH-associated genes as well as in new candidate genes.


Subject(s)
Arthritis, Juvenile/genetics , Lymphohistiocytosis, Hemophagocytic/genetics , Macrophage Activation Syndrome/genetics , Adolescent , Arthritis, Juvenile/complications , Child , Child, Preschool , Exome , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant , Macrophage Activation Syndrome/complications , Male , Mutation , Parents , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...