Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Clin Immunol ; 42(4): 851-858, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35305204

RESUMEN

Hematopoietic stem cell transplantation and gene therapy are the only curative therapies for severe combined immunodeficiency (SCID). In patients lacking a matched donor, TCRαß/CD19-depleted haploidentical family donor transplant (TCRαß-HaploSCT) is a promising strategy. Conditioned transplant in SCID correlates to better myeloid chimerism and reduced immunoglobulin dependency. We studied transplant outcome in SCID infants according to donor type, specifically TCRαß-HaploSCT, and conditioning, through retrospective cohort analysis of 52 consecutive infants with SCID transplanted between 2013 and 2020. Median age at transplant was 5.1 months (range, 0.8-16.6). Donors were TCRαß-HaploSCT (n = 16, 31.4%), matched family donor (MFD, n = 15, 29.4%), matched unrelated donor (MUD, n = 9, 17.6%), and matched unrelated cord blood (CB, n = 11, 21.6%). Forty-one (80%) received fludarabine/treosulfan-based conditioning, 3 (6%) had alemtuzumab only, and 7 (14%) received unconditioned infusions. For conditioned transplants (n = 41), 3-year overall survival was 91% (95% confidence interval, 52-99%) for TCRαß-HaploSCT, 80% (41-98%) for MFD, 87% (36-98%) for MUD, and 89% (43-98%) for CB (p = 0.89). Cumulative incidence of grade II-IV acute graft-versus-host disease was 11% (2-79%) after TCRαß-HaploSCT, 0 after MFD, 29% (7-100%) after MUD, and 11% (2-79%) after CB (p = 0.10). 9/10 patients who received alemtuzumab-only or unconditioned transplants survived. Myeloid chimerism was higher following conditioning (median 47%, range 0-100%) versus unconditioned transplant (median 3%, 0-9%) (p < 0.001), as was the proportion of immunoglobulin-free long-term survivors (n = 29/36, 81% vs n = 4/9, 54%) (p < 0.001). TCRαß-HaploSCT has comparable outcome to MUD and is a promising alternative donor strategy for infants with SCID lacking MFD. This study confirms that conditioned transplant offers better myeloid chimerism and immunoglobulin freedom in long-term survivors.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Alemtuzumab , Enfermedad Injerto contra Huésped/etiología , Humanos , Lactante , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/cirugía , Acondicionamiento Pretrasplante , Donante no Emparentado
2.
J Clin Immunol ; 42(4): 819-826, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35288820

RESUMEN

Current treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID) includes enzyme replacement therapy (ERT), allogeneic hematopoietic stem cell transplant (HSCT), or ex vivo corrected autologous hematopoietic stem cell gene therapy. Historic data show HSCT survival is superior using unconditioned matched sibling and family compared to matched unrelated and haploidentical donors. Recent improvement in HSCT outcomes prompted us to retrospectively examine HSCT survival and long-term graft function in ADA-SCID transplanted at our center. Thirty-three ADA-deficient patients received HSCT between 1989 and 2020, with follow-up data to January 2021. Chemotherapy conditioning regimens were defined as myeloablative (MAC-busulfan/cyclophosphamide), reduced-toxicity myeloablative (RT-MAC-treosulfan-based, since 2007), or no conditioning. Serotherapy used included alemtuzumab (with or without other conditioning agents) or antithymocyte globulin (ATG). ERT was introduced routinely in 2010 until commencement of conditioning. Median age at HSCT was 3.2 (0.8-99.8) months. Twenty-one (63.6%) received stem cells from unrelated or haploidentical donors. Seventeen (51.5%) received chemotherapy conditioning and 16 (48.5%) received alemtuzumab. Median follow-up was 7.5 (0.8-25.0) years. Overall survival (OS) and event-free survival (EFS) at 8 years were 90.9% (95% CI: 79.7-100.0%) and 79% (55-91%), respectively. OS after 2007 (n = 21) was 100% vs 75% before 2007 (n = 12) (p = 0.02). Three (9.1%) died after HSCT: two from multiorgan failure and one from unexplained encephalopathy. There were no deaths after 2007, among those who received ERT and treosulfan-based conditioning pre-HSCT. Ten (30.3%) developed acute GvDH (3 grade II, 2 grade III); no chronic GvHD was observed. In the modern era, conditioned HSCT with MUD has a favorable outcome for ADA-deficient patients.


Asunto(s)
Adenosina Desaminasa , Agammaglobulinemia , Terapia de Reemplazo Enzimático , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Adenosina Desaminasa/genética , Agammaglobulinemia/cirugía , Alemtuzumab/uso terapéutico , Terapia de Reemplazo Enzimático/métodos , Terapia Genética/métodos , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/cirugía , Acondicionamiento Pretrasplante
3.
J Clin Immunol ; 39(8): 753-761, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31432442

RESUMEN

Severe combined immune deficiency (SCID) is caused by an array of genetic disorders resulting in a diminished adaptive immune system due to impaired T lymphocytes. In these patients, active infection at the time of hematopoietic transplantation has been shown to increase morbidity and mortality. To prevent transmission of infections in SCID patients, standardized infection control precautions should be implemented. An online survey regarding SCID-specific protocols was distributed through several immunodeficiency organizations. Seventy-three responses were obtained, with the majority (55%) of responses from the USA, 15% from Canada, and the remainder from 12 other countries. Only 50% of respondents had a SCID-specific infection control protocol at their center, and while a majority of these centers had training for physicians, a small minority had training for other healthcare workers such as nursing and housekeeping staff. Significant variability of infection control practices, such as in-patient precautions, required personal protective equipment (PPE), diet restrictions, visitor precautions and discharge criteria, was found between different treatment centers. There is a paucity of evidence-based data regarding the safest environment to prevent infection in SCID patients. Institutional protocols may have significant impact on infection risk, survival, family well-being, child development and cost of care. From these results, it is evident that further multi-center research is required to determine the safest and healthiest environment for these children, so that evidence-based infection control protocols for patients with SCID can be developed.


Asunto(s)
Infección Hospitalaria/prevención & control , Medicina Basada en la Evidencia/estadística & datos numéricos , Control de Infecciones/estadística & datos numéricos , Pautas de la Práctica en Medicina/estadística & datos numéricos , Inmunodeficiencia Combinada Grave/inmunología , Lactancia Materna , Cuidadores/normas , Protocolos Clínicos , Infección Hospitalaria/inmunología , Medicina Basada en la Evidencia/instrumentación , Medicina Basada en la Evidencia/organización & administración , Medicina Basada en la Evidencia/normas , Trasplante de Células Madre Hematopoyéticas/normas , Humanos , Higiene/normas , Lactante , Recién Nacido , Control de Infecciones/instrumentación , Control de Infecciones/organización & administración , Control de Infecciones/normas , Educación del Paciente como Asunto , Equipo de Protección Personal/normas , Pautas de la Práctica en Medicina/organización & administración , Pautas de la Práctica en Medicina/normas , Inmunodeficiencia Combinada Grave/cirugía , Encuestas y Cuestionarios/estadística & datos numéricos
4.
Wound Repair Regen ; 27(4): 426-430, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843296

RESUMEN

Transplantation of human xenografts onto immunocompromised mice is a powerful research tool for studying wound healing. However, differences in healing between humans and mice and their small size limit this model. We determined whether human cadaver skin xenografts transplanted onto pigs with severe combined immune deficiency (SCID) would survive and not be rejected. Meshed (1:1.5), cryopreserved human cadaver skin was transplanted onto 10 partial thickness dermatome wounds in each of two normal domestic pigs and two SCID pigs. Autografts (n = 2/animal) from the four animals were used as controls. In normal pigs, all autografts were engrafted and healed with a minimal, if any, inflammation and scarring. All human xenografts were rejected by the normal pigs within 5-11 days and associated with an intense T-cell inflammatory response. In contrast, both autografts and xenografts were engrafted and survived the 28-day study in the SCID pigs with a minimal inflammation and no gross scarring.


Asunto(s)
Cadáver , Supervivencia de Injerto/fisiología , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/fisiopatología , Trasplante de Piel , Trasplante Heterólogo , Animales , Modelos Animales de Enfermedad , Supervivencia de Injerto/inmunología , Humanos , Inmunohistoquímica , Prueba de Estudio Conceptual , Inmunodeficiencia Combinada Grave/cirugía , Porcinos , Cicatrización de Heridas/inmunología , Cicatrización de Heridas/fisiología
5.
J Paediatr Child Health ; 53(8): 766-770, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28513891

RESUMEN

AIM: To examine the long-term follow-up and health outcomes of patients who have undergone haematopoietic stem cell transplant (HSCT) for severe combined immunodeficiency (SCID). METHODS: Through a structured questionnaire, we examined follow-up arrangements and long-term health outcomes in 22 children who have had a successful HSCT for SCID during the period of 1984-2012 at the Sydney Children's Hospital, Sydney, Australia. RESULTS: Most children considered themselves healthy and 'cured' from SCID. Whilst many children enjoy relatively good bio-social health outcomes, specific negative health outcomes and absenteeism from school were perceived negatively. Two-thirds of children see their general practitioner or specialist regularly; however, there did not appear to be consistency with the nature of this follow-up. CONCLUSION: The findings from our study highlight the complex bio-psychosocial health needs of post-HSCT SCID children and encourage SCID centres to consider a multidisciplinary approach to their follow-up. Further studies into the determinants of patients' perceptions of their health are needed.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Evaluación del Resultado de la Atención al Paciente , Inmunodeficiencia Combinada Grave/cirugía , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Nueva Gales del Sur , Evaluación de Resultado en la Atención de Salud , Inmunodeficiencia Combinada Grave/diagnóstico , Encuestas y Cuestionarios
6.
Pediatr Transplant ; 20(7): 888-897, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27241476

RESUMEN

Chronic IS is associated with significant morbidity in transplant recipients. Moreover, IS does not prevent chronic graft failure frequently. Allograft immune tolerance in LT can be induced by complete donor chimerism through allogenic HSCT combined with identical LDLT. This approach may exempt patients from chronic lifelong IS. However, it is unclear whether its benefits justify its risks. Here, we present three cases from our institution and analyze seven additional reports of children treated with HSCT/LDLT, all receiving HSCT due to hemato-oncological indications. In eight of 10 cases, donor macrochimerism resulted in allograft tolerance. Nine patients survived. One patient died due to fulminant ADV infection. Further complications were GvHD (n = 3) and bone marrow failure (n = 2). In conclusion, donor-specific allograft tolerance can be achieved by identical-donor HSCT/LDLT. However, at present, this approach should generally be limited to selected indications due to a potentially unfavorable risk-benefit ratio. Novel toxicity-reduced conditioning protocols for HSCT/LDLT in the absence of malignant or non-hepatic disease may prove to be a sufficiently safe approach for inducing graft tolerance in children receiving a LDLT in the future. This concept may reduce the burden of lifelong IS.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Hígado/métodos , Donadores Vivos , Aloinjertos , Preescolar , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/etiología , Hepatoblastoma/cirugía , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Inmunosupresores/uso terapéutico , Lactante , Neoplasias Hepáticas/cirugía , Pediatría , Calidad de Vida , Inmunodeficiencia Combinada Grave/cirugía , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento
7.
Clin Exp Immunol ; 176(1): 23-36, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24261689

RESUMEN

Fetal and neonatal alloimmune thrombocytopenia (FNAIT) occurs most frequently when human platelet antigen (HPA)-1a-positive fetal platelets are destroyed by maternal HPA-1a immunoglobulin (Ig)G antibodies. Pregnancies at risk are treated by administration of high-dose intravenous Ig (IVIG) to women, but this is expensive and often not well tolerated. Peptide immunotherapy may be effective for ameliorating some allergic and autoimmune diseases. The HPA-1a/1b polymorphism is Leu/Pro33 on ß3 integrin (CD61), and the anti-HPA-1a response is restricted to HPA-1b1b and HLA-DRB3*0101-positive pregnant women with an HPA-1a-positive fetus. We investigated whether or not HPA-1a antigen-specific peptides that formed the T cell epitope could reduce IgG anti-HPA-1a responses, using a mouse model we had developed previously. Peripheral blood mononuclear cells (PBMC) in blood donations from HPA-1a-immunized women were injected intraperitoneally (i.p.) into severe combined immunodeficient (SCID) mice with peptides and HPA-1a-positive platelets. Human anti-HPA-1a in murine plasma was quantitated at intervals up to 15 weeks. HPA-1a-specific T cells in PBMC were identified by proliferation assays. Using PBMC of three donors who had little T cell reactivity to HPA-1a peptides in vitro, stimulation of anti-HPA-1a responses by these peptides occurred in vivo. However, with a second donation from one of these women which, uniquely, had high HPA-1a-specific T cell proliferation in vitro, marked suppression of the anti-HPA-1a response by HPA-1a peptides occurred in vivo. HPA-1a peptide immunotherapy in this model depended upon reactivation of HPA-1a T cell responses in the donor. For FNAIT, we suggest that administration of antigen-specific peptides to pregnant women might cause either enhancement or reduction of pathogenic antibodies.


Asunto(s)
Antígenos de Plaqueta Humana/inmunología , Péptidos/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Plaqueta Humana/química , Antígenos de Plaqueta Humana/genética , Plaquetas/inmunología , Células Cultivadas , Citocinas/sangre , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Genotipo , Cadenas HLA-DRB3/genética , Cadenas HLA-DRB3/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Recién Nacido , Integrina beta3 , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Datos de Secuencia Molecular , Transfusión de Plaquetas , Embarazo , Inmunodeficiencia Combinada Grave/sangre , Inmunodeficiencia Combinada Grave/cirugía , Trombocitopenia Neonatal Aloinmune/sangre , Trombocitopenia Neonatal Aloinmune/inmunología , Trasplante Heterólogo
8.
Blood ; 119(12): 2949-55, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22308292

RESUMEN

Pediatric patients with SCID constitute medical emergencies. In the absence of an HLA-identical hematopoietic stem cell (HSC) donor, mismatched related-donor transplantation (MMRDT) or unrelated-donor umbilical cord blood transplantation (UCBT) are valuable treatment options. To help transplantation centers choose the best treatment option, we retrospectively compared outcomes after 175 MMRDTs and 74 UCBTs in patients with SCID or Omenn syndrome. Median follow-up time was 83 months and 58 months for UCBT and MMRDT, respectively. Most UCB recipients received a myeloablative conditioning regimen; most MMRDT recipients did not. UCB recipients presented a higher frequency of complete donor chimerism (P = .04) and faster total lymphocyte count recovery (P = .04) without any statistically significance with the preparative regimen they received. The MMRDT and UCBT groups did not differ in terms of T-cell engraftment, CD4(+) and CD3(+) cell recoveries, while Ig replacement therapy was discontinued sooner after UCBT (adjusted P = .02). There was a trend toward a greater incidence of grades II-IV acute GVHD (P = .06) and more chronic GVHD (P = .03) after UCBT. The estimated 5-year overall survival rates were 62% ± 4% after MMRDT and 57% ± 6% after UCBT. For children with SCID and no HLA-identical sibling donor, both UCBT and MMRDT represent available HSC sources for transplantation with quite similar outcomes.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Enfermedad Injerto contra Huésped/epidemiología , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/cirugía , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Trasplante de Células Madre de Sangre del Cordón Umbilical/mortalidad , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Incidencia , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/mortalidad , Acondicionamiento Pretrasplante/métodos , Resultado del Tratamiento
9.
Br J Haematol ; 160(2): 146-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23167301

RESUMEN

Severe combined immunodeficiencies (SCIDs) correspond to the most severe form of primary immunodeficiency. The extreme severity of the clinical presentation in SCID has legitimately led physicians to consider these conditions as medical emergencies. Hundreds of patients worldwide have undergone allogeneic haematopoietic stem cell transplantation (HCST) in the last 40 years. The complete absence of the T cell compartment in SCID prompted the development (starting in the early 1980s) of haploidentical, parental HSCT for the many patients who do not have a human leucocyte antigen (HLA)-identical sibling. Despite the undeniable progress made in this field over recent years, the long-lasting immunodeficiency that follows partially HLA-incompatible transplantation is still responsible for a mortality rate of 30% at one year post-transplantation. New approaches for reconstituting T cell compartments more rapidly are under intense preclinical development and are discussed herein.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/cirugía , Trasplante de Células Madre de Sangre del Cordón Umbilical , Citocinas/inmunología , Europa (Continente)/epidemiología , Terapia Genética , Rechazo de Injerto/inmunología , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Lactante , Recién Nacido , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Subgrupos Linfocitarios/trasplante , Transfusión de Linfocitos , Linfopoyesis , Tamizaje Neonatal , Inmunodeficiencia Combinada Grave/clasificación , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/embriología , Timo/inmunología , Timo/patología , Donantes de Tejidos , Trasplante Homólogo , Resultado del Tratamiento , Estados Unidos/epidemiología
10.
J Clin Immunol ; 33(1): 96-110, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23001410

RESUMEN

PURPOSE: Severe combined immunodeficiency (SCID) is a syndrome of diverse genetic cause characterized by profound deficiencies of T, B and sometimes NK cell function. Non-ablative HLA-identical or rigorously T cell-depleted haploidentical parental bone marrow transplantation (BMT) results in thymus-dependent genetically donor T cell development in the recipients, leading to a high rate of long-term survival. However, the development of B cell function has been more problematic. We report here results of analyses of B cell function in 125 SCID recipients prior to and long-term after non-ablative BMT, according to their molecular type. METHODS: Studies included blood immunoglobulin measurements; antibody titers to standard vaccines, blood group antigens and bacteriophage Φ X 174; flow cytometry to examine for markers of immaturity, memory, switched memory B cells and BAFF receptor expression; B cell chimerism; B cell spectratyping; and B cell proliferation. RESULTS: The results showed that B cell chimerism was not required for normal B cell function in IL7Rα-Def, ADA-Def and CD3-Def SCIDs. In X-linked-SCID, Jak3-Def SCID and those with V-D-J recombination defects, donor B cell chimerism was necessary for B cell function to develop. CONCLUSION: The most important factor determining whether B cell function develops in SCID T cell chimeras is the underlying molecular defect. In some types, host B cells function normally. In those molecular types where host B cell function did not develop, donor B cell chimerism was necessary to achieve B cell function. 236 words.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/trasplante , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/patología , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Adulto , Subgrupos de Linfocitos B/patología , Trasplante de Médula Ósea/métodos , Transformación Celular Viral/inmunología , Células Cultivadas , Niño , Femenino , Humanos , Inmunofenotipificación , Lactante , Células Jurkat , Depleción Linfocítica , Transfusión de Linfocitos/métodos , Masculino , Periodo Posoperatorio , Quimera por Radiación/inmunología , Inmunodeficiencia Combinada Grave/cirugía , Cariotipificación Espectral , Subgrupos de Linfocitos T/patología , Quimera por Trasplante/inmunología , Células Tumorales Cultivadas
11.
Blood ; 117(2): 688-96, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20978268

RESUMEN

FOXN1 deficiency is a primary immunodeficiency characterized by athymia, alopecia totalis, and nail dystrophy. Two infants with FOXN1 deficiency were transplanted with cultured postnatal thymus tissue. Subject 1 presented with disseminated Bacillus Calmette-Guérin infection and oligoclonal T cells with no naive markers. Subject 2 had respiratory failure, human herpes virus 6 infection, cytopenias, and no circulating T cells. The subjects were given thymus transplants at 14 and 9 months of life, respectively. Subject 1 received immunosuppression before and for 10 months after transplantation. With follow up of 4.9 and 2.9 years, subjects 1 and 2 are well without infectious complications. The pretransplantation mycobacterial disease in subject 1 and cytopenias in subject 2 resolved. Subject 2 developed autoimmune thyroid disease 1.6 years after transplantation. Both subjects developed functional immunity. Subjects 1 and 2 have 1053/mm(3) and 1232/mm(3) CD3(+) cells, 647/mm(3) and 868/mm(3) CD4(+) T cells, 213/mm(3) and 425/mm(3) naive CD4(+) T cells, and 10 200 and 5700 T-cell receptor rearrangement excision circles per 100 000 CD3(+) cells, respectively. They have normal CD4 T-cell receptor ß variable repertoires. Both subjects developed antigen-specific proliferative responses and have discontinued immunoglobulin replacement. In summary, thymus transplantation led to T-cell reconstitution and function in these FOXN1 deficient infants.


Asunto(s)
Factores de Transcripción Forkhead/deficiencia , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/fisiopatología , Inmunodeficiencia Combinada Grave/cirugía , Timo/trasplante , Separación Celular , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Lactante , Masculino
12.
J Pediatr Hematol Oncol ; 35(8): e335-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23389499

RESUMEN

We report a RAG2-deficient patient with severe combined immunodeficiency and hemophagocytic bone marrow aplasia with plasma cells after a nonconditioned transplantation from a fully matched sibling. After engraftment, disseminated BCGosis appeared because of graft versus host disease prophylaxis. On the 55th day, eosinophilia, neutropenia, and thrombocytopenia developed. Aplasia, hemophagocytic histiocytes, and plasma cells were found on his bone marrow with very high level of serum immunoglobulin E. We could not discriminate exactly whether BCGosis or alloimmune response is the cause of hemophagocytic aplasia with plasma cells. Despite the second hematopoietic stem cell transplantation with a reduced intensity conditioning regime, his marrow aplasia did not recover and he died. This case suggests that BCGosis might be associated with hemophagocytic marrow aplasia with plasma cells in an alloimmune reaction.


Asunto(s)
Vacuna BCG/efectos adversos , Enfermedades de la Médula Ósea/etiología , Proteínas de Unión al ADN/genética , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfohistiocitosis Hemofagocítica/etiología , Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/cirugía , Vacuna BCG/inmunología , Enfermedades de la Médula Ósea/patología , Humanos , Lactante , Linfohistiocitosis Hemofagocítica/patología , Masculino , Células Plasmáticas/patología , Inmunodeficiencia Combinada Grave/genética , Acondicionamiento Pretrasplante , Trasplante Homólogo/efectos adversos
13.
Biol Blood Marrow Transplant ; 18(12): 1911-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22842333

RESUMEN

This study analyzes the hematopoietic cell transplantation experience in patients with immune deficiency at a single institution. The objective is to comprehensively evaluate the short-term and long-term outcomes with various preparative regimens, donor grafts, and ex vivo manipulations to identify transplantation approaches that most likely favor early donor immune competency without generating excessive toxicity. Clinical outcomes were evaluated in 52 consecutive patients with immune deficiencies. Thirty-seven of the 52 patients (71%) survived with attenuation of their underlying disease. The use of a melphalan-based reduced-intensity conditioning preparative regimen and immunomagnetic CD3(+) T cell depletion techniques (when T cell depletion was indicated) were associated with improved event-free survival. Survivors who received a preparative regimen other than a melphalan-based reduced-intensity regimen suffered from therapy-related morbidities or chronic/recurrent infections. Our findings indicate that melphalan-based reduced-intensity conditioning regimens and immunomagnetic CD3(+) T cell depletion limit therapy-related toxicity, and demonstrate promising results for the early establishment of donor immune competency.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Depleción Linfocítica/métodos , Inmunodeficiencia Combinada Grave/cirugía , Linfocitos T/inmunología , Adolescente , Adulto , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/citología , Trasplante Homólogo , Adulto Joven
14.
Pediatr Transplant ; 16(5): E167-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21535335

RESUMEN

SCID affects T and B cell differentiation and functions, presenting with severe opportunistic infections in the early postnatal period. It is fatal unless stem cell transplantation is performed. RS SCID forms are caused by defects in the NHEJ pathway, the enzymatic process required for the repair of DNA double-strand breaks. Cernunnos-XLF defect is one of the defects in this pathway. Here, we present two patients with Cernunnos-XLF defect, both having microcephaly, prominent growth retardation, and T-B-NK+SCID, one of whom had AHA. These patients received hematopoietic stem cells from HLA identical related donor without conditioning regimen and recovered without any complication. Now, both of the patients are well and alive seven and one yr after transplantation, respectively. A remarkable observation was the severe diarrhea that occurred in both patients soon after transplantation.


Asunto(s)
Trasplante de Médula Ósea , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/cirugía , Enzimas Reparadoras del ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Femenino , Marcadores Genéticos , Humanos , Lactante , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo
15.
Pediatr Transplant ; 16(2): E43-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21108710

RESUMEN

Omenn's syndrome is a rare inherited variant of SCID. It is inevitably fatal, unless treated by bone marrow or stem cell transplantation. However, treatment-related complications and graft rejection are major obstacles to the success of transplantation. In this report, we describe an eight-month-old baby with Omenn's syndrome and disseminated BCGosis, who underwent allogenic BMT from his HLA-matched sister using anti-GVHD prophylaxis but without a conditioning regimen. Ten days after BMT, he developed acute GVHD involving the skin only (grade 1) with a flare of local BCGosis. Engraftment of 55% was demonstrated by FISH as early as the 11th day post-BMT. On day +48 post-BMT, he developed GVHD that was complicated by refractory pancytopenia, resistant to three doses of rituximab. Patient was re-transplanted (from the same donor) after receiving fludarabine and ATG, with successful engraftment and complete recovery from pancytopenia.


Asunto(s)
Trasplante de Médula Ósea , Pancitopenia/cirugía , Trasplante de Células Madre de Sangre Periférica , Complicaciones Posoperatorias/cirugía , Inmunodeficiencia Combinada Grave/cirugía , Acondicionamiento Pretrasplante , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Lactante , Masculino , Mycobacterium bovis , Pancitopenia/diagnóstico , Pancitopenia/etiología , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/prevención & control , Reoperación , Inmunodeficiencia Combinada Grave/complicaciones , Inmunodeficiencia Combinada Grave/diagnóstico , Tuberculosis/diagnóstico , Tuberculosis/etiología
16.
Pediatr Transplant ; 16(5): 451-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22594916

RESUMEN

SCID is characterized by profound deficiencies of T and B lymphocytes. HSCT is the only curative treatment for children with SCID. The clinical characteristics and outcome of 30 HLA-haploidentical transplantations in 18 patients (15 SCID, two Omenn syndrome, and one MHC Class II deficiency) are reported here. The age of patients at diagnosis ranged from one and half to nine months (median: four months). The median time was one month between the diagnosis and the time of the initial transplantation. Infused CD34+ stem cell dose was ranged between 7 and 94.2 × 10(6) /kg. Nine of 18 patients were found to be positive for CMV antigenemia at diagnosis; therefore, none of them received a conditioning regimen. The most common complication was graft failure (61%), so repeated transplantations (two to four) were performed in seven patients. The mean time of lymphoid engraftment was 17.5 days (median: 16, range: 11-29 days). Ten of 15 SCID (67%) patients survived with a stable complete donor chimerism. However, all three non-SCID patients died. In conclusion, in the absence of a matched family donor, HLA-haploidentical transplantation from parental donors represents a readily available treatment option especially for patients with SCID, offering a high chance of cure.


Asunto(s)
Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Histocompatibilidad , Inmunodeficiencia Combinada Grave/cirugía , Selección de Donante , Femenino , Estudios de Seguimiento , Rechazo de Injerto/epidemiología , Humanos , Lactante , Masculino , Padres , Complicaciones Posoperatorias/epidemiología , Reoperación/estadística & datos numéricos , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/mortalidad , Tasa de Supervivencia , Resultado del Tratamiento
17.
J Allergy Clin Immunol ; 128(5): 1050-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21757226

RESUMEN

BACKGROUND: CD3δ deficiency is a fatal form of severe combined immunodeficiency that can be cured by hematopoietic stem cell transplantation (HSCT). The presence of a thymus loaded with T-cell progenitors in patients with CD3δ deficiency may require special considerations in choosing the regimen of conditioning and the type of HSCT. OBJECTIVES: To study the outcome of CD3δ deficiency by using various modalities of stem cell transplantation. METHODS: We analyzed data on 13 patients with CD3δ deficiency who underwent HSCT in 7 centers. HSCT was performed by using different sources of donor stem cells as well as various conditioning regimens. RESULTS: One patient received stem cells from a matched related donor and survived after a second transplant, needing substantial conditioning in order to engraft. Only 2 of 7 other patients who received a mismatched related donor transplant survived; 2 of them had no conditioning, whereas the others received various combinations of conditioning regimens. Engraftment of T cells in the survivors appears incomplete. Three other patients who received stem cells from a matched unrelated donor survived and enjoyed full immune reconstitution. Two patients received unrelated cord blood without conditioning. One of them has had a partial but stable engraftment, whereas the other engrafted well but is only 12 months after HSCT. We also report here for the first time that patients with CD3δ deficiency can present with typical features of Omenn syndrome. CONCLUSIONS: HSCT is a successful treatment for patients with CD3δ deficiency. The small number of patients in this report prevents definitive statements on the importance of survival factors, but several are suggested: (1) HLA-matched donor transplants are associated with superior reconstitution and survival than are mismatched donor transplants; (2) substantial conditioning appears necessary; and (3) early diagnosis and absence of opportunistic infections may affect outcome.


Asunto(s)
Complejo CD3/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/cirugía , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Complicaciones Posoperatorias/epidemiología , Acondicionamiento Pretrasplante/métodos
18.
Blood ; 113(17): 4114-24, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19168787

RESUMEN

Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for severe combined immunodeficiency (SCID). Detailed assessment of the long-term outcome of HSCT, ie, the occurrence of clinical events and the quality and stability of immune reconstitution, is now required. We performed a single-center retrospective analysis of the long-term outcome of HSCT in 90-patient cohort followed for between 2 and 34 years (median, 14 years). Clinical events and immune reconstitution data were collected. Almost half the patients have experienced one or more significant clinical events, including persistent chronic graft-versus-host disease (GVHD), autoimmune and inflammatory manifestations, opportunistic and nonopportunistic infections, chronic human papilloma virus (HPV) infections, and a requirement for nutritional support. With the notable exception of severe HPV infection, these complications tend to become less common 15 years later after HSCT. A multivariate analysis showed that the occurrence of these events correlated with non-genoidentical donors, diagnosis of Artemis SCID, and quality of immune reconstitution. In most cases, HSCT enables long-term survival with infrequent sequelae. However, the occurrence of relatively late-onset complications is a concern that requires specific means of prevention and justifies careful patient follow-up.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/cirugía , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/psicología , Tasa de Supervivencia , Factores de Tiempo , Resultado del Tratamiento
19.
J Paediatr Child Health ; 47(8): 565-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21843190

RESUMEN

We present a case of a 2-month-old boy with partially treated meningitis and suspected Pneumocystis carinii pneumonia. A full blood count revealed profound lymphopenia. The child was diagnosed with adenosine deaminase deficiency, a rare cause of severe combined immunodeficiency (SCID). SCID is an immunological emergency and must be considered in any lymphopaenic infant with opportunistic infection. We discuss adenosine deaminase-deficient SCID, which can involve multiple systems and in which other treatment options apart from bone marrow transplant are available.


Asunto(s)
Linfopenia/complicaciones , Inmunodeficiencia Combinada Grave/complicaciones , Inmunodeficiencia Combinada Grave/diagnóstico , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Linfopenia/sangre , Linfopenia/diagnóstico , Masculino , Inmunodeficiencia Combinada Grave/sangre , Inmunodeficiencia Combinada Grave/cirugía , Resultado del Tratamiento
20.
Front Immunol ; 12: 721917, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095830

RESUMEN

Congenital athymia can present with severe T cell lymphopenia (TCL) in the newborn period, which can be detected by decreased T cell receptor excision circles (TRECs) on newborn screening (NBS). The most common thymic stromal defect causing selective TCL is 22q11.2 deletion syndrome (22q11.2DS). T-box transcription factor 1 (TBX1), present on chromosome 22, is responsible for thymic epithelial development. Single variants in TBX1 causing haploinsufficiency cause a clinical syndrome that mimics 22q11.2DS. Definitive therapy for congenital athymia is allogeneic thymic transplantation. However, universal availability of such therapy is limited. We present a patient with early diagnosis of congenital athymia due to TBX1 haploinsufficiency. While evaluating for thymic transplantation, she developed Omenn Syndrome (OS) and life-threatening adenoviremia. Despite treatment with anti-virals and cytotoxic T lymphocytes (CTLs), life threatening adenoviremia persisted. Given the imminent need for rapid establishment of T cell immunity and viral clearance, the patient underwent an unmanipulated matched sibling donor (MSD) hematopoietic cell transplant (HCT), ultimately achieving post-thymic donor-derived engraftment, viral clearance, and immune reconstitution. This case illustrates that because of the slower immune recovery that occurs following thymus transplantation and the restricted availability of thymus transplantation globally, clinicians may consider CTL therapy and HCT to treat congenital athymia patients with severe infections.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Proteínas de Dominio T Box/genética , Timo/anomalías , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Síndromes de Inmunodeficiencia/cirugía , Recién Nacido , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/cirugía , Hermanos , Timo/cirugía
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda