Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 143(2): 118-123, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-37647647

RESUMEN

ABSTRACT: CD19-negative relapse is a leading cause of treatment failure after chimeric antigen receptor (CAR) T-cell therapy for acute lymphoblastic leukemia. We investigated a CAR T-cell product targeting CD19 and CD22 generated by lentiviral cotransduction with vectors encoding our previously described fast-off rate CD19 CAR (AUTO1) combined with a novel CD22 CAR capable of effective signaling at low antigen density. Twelve patients with advanced B-cell acute lymphoblastic leukemia were treated (CARPALL [Immunotherapy with CD19/22 CAR Redirected T Cells for High Risk/Relapsed Paediatric CD19+ and/or CD22+ Acute Lymphoblastic Leukaemia] study, NCT02443831), a third of whom had failed prior licensed CAR therapy. Toxicity was similar to that of AUTO1 alone, with no cases of severe cytokine release syndrome. Of 12 patients, 10 (83%) achieved a measurable residual disease (MRD)-negative complete remission at 2 months after infusion. Of 10 responding patients, 5 had emergence of MRD (n = 2) or relapse (n = 3) with CD19- and CD22-expressing disease associated with loss of CAR T-cell persistence. With a median follow-up of 8.7 months, there were no cases of relapse due to antigen-negative escape. Overall survival was 75% (95% confidence interval [CI], 41%-91%) at 6 and 12 months. The 6- and 12-month event-free survival rates were 75% (95% CI, 41%-91%) and 60% (95% CI, 23%-84%), respectively. These data suggest dual targeting with cotransduction may prevent antigen-negative relapse after CAR T-cell therapy.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Humanos , Niño , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos/genética , Recurrencia , Antígenos CD19 , Linfocitos T , Lectina 2 Similar a Ig de Unión al Ácido Siálico
2.
N Engl J Med ; 386(5): 415-427, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34891223

RESUMEN

BACKGROUND: Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent ß-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the ß-globin (ßA-T87Q) gene. METHODS: In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent ß-thalassemia and a non-ß0/ß0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS: A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS: Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-ß0/ß0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).


Asunto(s)
Productos Biológicos/uso terapéutico , Terapia Genética/métodos , Globinas beta/genética , Talasemia beta/terapia , Adolescente , Adulto , Productos Biológicos/efectos adversos , Busulfano/uso terapéutico , Niño , Transfusión de Eritrocitos/efectos adversos , Eritropoyesis , Femenino , Vectores Genéticos , Genotipo , Hemoglobinas/análisis , Humanos , Sobrecarga de Hierro/prevención & control , Lentivirus/genética , Masculino , Persona de Mediana Edad , Agonistas Mieloablativos/uso terapéutico , Talasemia beta/sangre , Talasemia beta/genética
3.
Mol Ther ; 32(6): 1672-1686, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38549377

RESUMEN

Stem cell gene therapy and hematopoietic stem cell transplantation (SCT) require conditioning to ablate the recipient's hematopoietic stem cells (HSCs) and create a niche for gene-corrected/donor HSCs. Conventional conditioning agents are non-specific, leading to off-target toxicities and resulting in significant morbidity and mortality. We developed tissue-specific anti-human CD45 antibody-drug conjugates (ADCs), using rat IgG2b anti-human CD45 antibody clones YTH24.5 and YTH54.12, conjugated to cytotoxic pyrrolobenzodiazepine (PBD) dimer payloads with cleavable (SG3249) or non-cleavable (SG3376) linkers. In vitro, these ADCs internalized to lysosomes for drug release, resulting in potent and specific killing of human CD45+ cells. In humanized NSG mice, the ADCs completely ablated human HSCs without toxicity to non-hematopoietic tissues, enabling successful engraftment of gene-modified autologous and allogeneic human HSCs. The ADCs also delayed leukemia onset and improved survival in CD45+ tumor models. These data provide proof of concept that conditioning with anti-human CD45-PBD ADCs allows engraftment of donor/gene-corrected HSCs with minimal toxicity to non-hematopoietic tissues. Our anti-CD45-PBDs or similar agents could potentially shift the paradigm in transplantation medicine that intensive chemo/radiotherapy is required for HSC engraftment after gene therapy and allogeneic SCT. Targeted conditioning both improve the safety and minimize late effects of these procedures, which would greatly increase their applicability.


Asunto(s)
Benzodiazepinas , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Inmunoconjugados , Antígenos Comunes de Leucocito , Animales , Humanos , Ratones , Inmunoconjugados/farmacología , Antígenos Comunes de Leucocito/metabolismo , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Benzodiazepinas/farmacología , Benzodiazepinas/química , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Ratas , Acondicionamiento Pretrasplante/métodos , Modelos Animales de Enfermedad , Anticuerpos Monoclonales/farmacología , Pirroles
4.
Br J Haematol ; 204(5): 1687-1696, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38488312

RESUMEN

The objective of this guideline, prepared by the ALL subgroup of the Advanced Cell Therapy Sub-Committee of BSBMTCT (British Society of Blood and Marrow Transplantation), is to provide healthcare professionals with practical guidance on the preparation of children and young adults with B-acute lymphoblastic leukaemia from the point of referral to that of admission for CAR T-cell treatment. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) nomenclature was used to evaluate the levels of evidence and to assess the strength of recommendations. The GRADE criteria can be found at http://www.gradeworkinggroup.org.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Niño , Inmunoterapia Adoptiva/métodos , Adulto Joven , Adolescente , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Adulto , Receptores Quiméricos de Antígenos/uso terapéutico
5.
Cytotherapy ; 25(1): 82-93, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36220712

RESUMEN

BACKGROUND AIMS: Delayed immune reconstitution is a major challenge after matched unrelated donor (MUD) stem cell transplant (SCT). In this randomized phase 2 multi-center trial, Adoptive Immunotherapy with CD25/71 allodepleted donor T cells to improve immunity after unrelated donor stem cell transplant (NCT01827579), the authors tested whether allodepleted donor T cells (ADTs) can safely be used to improve immune reconstitution after alemtuzumab-based MUD SCT for hematological malignancies. METHODS: Patients received standard of care or up to three escalating doses of ADTs generated through CD25+/CD71+ immunomagnetic depletion. The primary endpoint of the study was circulating CD3+ T-cell count at 4 months post-SCT. Twenty-one patients were treated, 13 in the ADT arm and eight in the control arm. RESULTS: The authors observed a trend toward improved CD3+ T-cell count at 4 months in the ADT arm versus the control arm (230/µL versus 145/µL, P = 0.18), and three ADT patients achieved normal CD3+ T-cell count at 4 months (>700/µL). The rates of significant graft-versus-host disease (GVHD) were comparable in both cohorts, with grade ≥2 acute GVHD in seven of 13 and four of eight patients and chronic GVHD in three of 13 and three of eight patients in the ADT and control arms, respectively. CONCLUSIONS: These data suggest that adoptive transfer of ADTs is safe, but that in the MUD setting the benefit in terms of T-cell reconstitution is limited. This approach may be of more use in the context of more rigorous T-cell depletion.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Linfocitos T , Donante no Emparentado , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inmunoterapia
6.
Br J Haematol ; 195(2): 249-255, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34431085

RESUMEN

Chronic active Epstein-Barr virus (CAEBV) disease is a rare condition characterised by persistent EBV infection in previously healthy individuals. Defective EBV genomes were found in East Asian patients with CAEBV. In the present study, we sequenced 14 blood EBV samples from three UK patients with CAEBV, comparing the results with saliva CAEBV samples and other conditions. We observed EBV deletions in blood, some of which may disrupt viral replication, but not saliva in CAEBV. Deletions were lost overtime after successful treatment. These findings are compatible with CAEBV being associated with the evolution and persistence of EBV+ haematological clones that are lost on successful treatment.


Asunto(s)
Infecciones por Virus de Epstein-Barr/sangre , Herpesvirus Humano 4/genética , Saliva/metabolismo , Eliminación de Secuencia/genética , Adolescente , Biomarcadores/análisis , Estudios de Casos y Controles , Niño , Preescolar , Enfermedad Crónica , Virus Defectuosos/genética , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/epidemiología , Asia Oriental/epidemiología , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , Trasplante de Células Madre de Sangre Periférica/métodos , Polimorfismo de Nucleótido Simple/genética , Rituximab/uso terapéutico , Resultado del Tratamiento , Replicación Viral/genética
7.
Br J Haematol ; 188(4): 560-569, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31566733

RESUMEN

Viral respiratory infections (VRIs) contribute to the morbidity and transplant-related mortality (TRM) after allogeneic haematopoietic stem cell transplantation (HSCT) and strategies to prevent and treat VRIs are warranted. We monitored VRIs before and after transplant in children undergoing allogeneic HSCT with nasopharyngeal aspirates (NPA) and assessed the impact on clinical outcome. Between 2007 and 2017, 585 children underwent 620 allogeneic HSCT procedures. Out of 75 patients with a positive NPA screen (12%), transplant was delayed in 25 cases (33%), while 53 children started conditioning with a VRI. Patients undergoing HSCT with a positive NPA screen had a significantly lower overall survival (54% vs. 79%) and increased TRM (26% vs. 7%) compared to patients with a negative NPA. Patients with a positive NPA who delayed transplant and cleared the virus before conditioning had improved overall survival (90%) and lower TRM (5%). Pre-HSCT positive NPA was the only significant risk factor for progression to a lower respiratory tract infection and was a major risk factor for TRM. Transplant delay, whenever feasible, in case of a positive NPA screen for VRIs can positively impact on survival of children undergoing HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Infecciones del Sistema Respiratorio/mortalidad , Acondicionamiento Pretrasplante , Virosis/mortalidad , Adolescente , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Estudios Retrospectivos , Tasa de Supervivencia , Factores de Tiempo
9.
Blood ; 124(16): 2514-22, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25185261

RESUMEN

Epstein-Barr virus (EBV)-associated posttransplant lymphoma (PTLD) is a major cause of morbidity/mortality after hematopoietic stem cell (SCT) or solid organ (SOT) transplant. Adoptive immunotherapy with EBV-specific cytotoxic lymphocytes (CTLs), although effective in SCT, is less successful after SOT where lifelong immunosuppression therapy is necessary. We have genetically engineered EBV-CTLs to render them resistant to calcineurin (CN) inhibitor FK506 through retroviral transfer of a calcineurin A mutant (CNA12). Here we examined whether or not FK506-resistant EBV-CTLs control EBV-driven tumor progression in the presence of immunosuppression in a xenogeneic mouse model. NOD/SCID/IL2rγ(null) mice bearing human B-cell lymphoma were injected with autologous CTLs transduced with either CNA12 or eGFP in the presence/absence of FK506. Adoptive transfer of autologous CNA12-CTLs induced dramatic lymphoma regression despite the presence of FK506, whereas eGFP-CTLs did not. CNA12-CTLs persisted longer, homed to the tumor, and expanded more than eGFP-CTLs in mice treated with FK506. Mice receiving CNA12-CTLs and treated with FK506 survived significantly longer than control-treated animals. Our results demonstrate that CNA12-CTL induce regression of EBV-associated tumors in vivo despite ongoing immunosuppression. Clinical application of this novel approach may enhance the efficacy of adoptive transfer of EBV-CTL in SOT patients developing PTLD without the need for reduction in immunosuppressive therapy.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Terapia Genética , Inmunoterapia Adoptiva , Linfoma/terapia , Linfoma/virología , Linfocitos T Citotóxicos/trasplante , Linfocitos T Citotóxicos/virología , Animales , Calcineurina/genética , Inhibidores de la Calcineurina/farmacología , Resistencia a Medicamentos , Ingeniería Genética/métodos , Terapia Genética/métodos , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Inmunosupresores/farmacología , Inmunoterapia Adoptiva/métodos , Linfoma/genética , Linfoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Tacrolimus/farmacología , Transducción Genética
10.
Blood ; 123(1): 126-32, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24184682

RESUMEN

In vivo T-cell depletion might contribute to the delayed immune reconstitution observed after unrelated umbilical cord blood transplantation (UCBT). We studied the impact of early, late, and no antithymocyte globulin (ATG) on immune reconstitution and outcome. One hundred twenty seven children receiving UCBT in London or Utrecht were divided into 3 groups: early ATG (days -9 to -5; n = 33), late ATG (days -5 to 0; n = 48), and no ATG (n = 46). The no-ATG group received mycophenolate mofetile + cyclosporin A as graft-versus-host disease (GVHD) prophylaxis, while the ATG groups received cyclosporin A + prednisone. End points studied were survival, immune recovery, infections, and GVHD. The probability of survival was similar in all groups: no ATG, 71% ± 8%; early ATG, 68% ± 9%; and late ATG, 61% ± 7%. CD3(+), CD4(+), and CD4(+)-naive T-cell counts were significantly higher (P < .001) in the no-ATG group at 1, 2, 3, 6, and 12 months post-UCBT. In the no-ATG group, significantly fewer viral reactivations (P = .021) were noted. A higher probability of severe acute GVHD (aGVHD; 31%) was found in the no-ATG group compared with 18% (P = .018) for early-ATG and 5% (P < .001) for late-ATG groups. This was not associated with more chronic GVHD (cGVHD).


Asunto(s)
Suero Antilinfocítico/metabolismo , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Sangre Fetal/citología , Adolescente , Suero Antilinfocítico/uso terapéutico , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inmunosupresores/uso terapéutico , Lactante , Masculino , Probabilidad , Inducción de Remisión , Estudios Retrospectivos , Factores de Riesgo , Acondicionamiento Pretrasplante/métodos , Resultado del Tratamiento , Adulto Joven
11.
Br J Haematol ; 171(4): 585-94, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26223288

RESUMEN

We explored the feasibility of unrelated donor haematopoietic stem cell transplant (HSCT) upfront without prior immunosuppressive therapy (IST) in paediatric idiopathic severe aplastic anaemia (SAA). This cohort was then compared to matched historical controls who had undergone first-line therapy with a matched sibling/family donor (MSD) HSCT (n = 87) or IST with horse antithymocyte globulin and ciclosporin (n = 58) or second-line therapy with unrelated donor HSCT post-failed IST (n = 24). The 2-year overall survival in the upfront cohort was 96 ± 4% compared to 91 ± 3% in the MSD controls (P = 0·30) and 94 ± 3% in the IST controls (P = 0·68) and 74 ± 9% in the unrelated donor HSCT post-IST failure controls (P = 0·02).The 2-year event-free survival in the upfront cohort was 92 ± 5% compared to 87 ± 4% in MSD controls (P = 0·37), 40 ± 7% in IST controls (P = 0·0001) and 74 ± 9% in the unrelated donor HSCT post-IST failure controls (n = 24) (P = 0·02). Outcomes for upfront-unrelated donor HSCT in paediatric idiopathic SAA were similar to MSD HSCT and superior to IST and unrelated donor HSCT post-IST failure. Front-line therapy with matched unrelated donor HSCT is a novel treatment approach and could be considered as first-line therapy in selected paediatric patients who lack a MSD.


Asunto(s)
Anemia Aplásica/terapia , Trasplante de Médula Ósea/estadística & datos numéricos , Trasplante de Células Madre de Sangre Periférica/estadística & datos numéricos , Infecciones por Adenoviridae/tratamiento farmacológico , Infecciones por Adenoviridae/epidemiología , Adolescente , Adulto , Anemia Aplásica/mortalidad , Suero Antilinfocítico , Transfusión Sanguínea/estadística & datos numéricos , Trasplante de Médula Ósea/efectos adversos , Estudios de Casos y Controles , Niño , Preescolar , Ciclosporina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/prevención & control , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/epidemiología , Histocompatibilidad , Humanos , Inmunosupresores/uso terapéutico , Lactante , Estimación de Kaplan-Meier , Tiempo de Internación/estadística & datos numéricos , Donadores Vivos , Masculino , Trasplante de Células Madre de Sangre Periférica/efectos adversos , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/epidemiología , Disfunción Primaria del Injerto/epidemiología , Calidad de Vida , Estudios Retrospectivos , Hermanos , Tasa de Supervivencia , Linfocitos T , Resultado del Tratamiento , Activación Viral , Adulto Joven
12.
Blood ; 122(23): 3749-58, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-24113871

RESUMEN

Severe combined immunodeficiency (SCID) arises from different genetic defects associated with lymphocyte development and function and presents with severe infections. Allogeneic hematopoietic stem cell transplantation is an extremely effective way of restoring immunity in these individuals. Numerous multicenter studies have identified the factors determining successful outcome, and survival for SCID has shown great improvement. Advances in understanding the genetic basis of disease also mean that we increasingly tailor transplant protocols to the specific SCID form. Wherever possible, we attempt to transplant SCID patients without the use of cytoreductive conditioning, but it is clear that this is only successful for specific SCID forms and, although survival is good, in specific patients there are ongoing humoral defects. We aim to use matched related and unrelated donors (including cord blood) whenever possible and have limited the use of mismatched haploidentical donors. The development of autologous hematopoietic stem cell gene therapy provides another treatment of the X-linked and adenosine deaminase-deficient forms of SCID, and we discuss how we have integrated gene therapy into our treatment strategy. These developments together with the advent of universal newborn screening for SCID should allow for a highly favorable outcome for this otherwise lethal condition.


Asunto(s)
Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/uso terapéutico , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Recién Nacido , Masculino , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Donantes de Tejidos , Acondicionamiento Pretrasplante
13.
Blood ; 121(6): 877-83, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23131490

RESUMEN

There have been no studies on patient outcome after allogeneic hematopoietic cell transplantation (HCT) in patients with X-linked inhibitor of apoptosis (XIAP) deficiency. To estimate the success of HCT, we conducted an international survey of transplantation outcomes. Data were reported for 19 patients. Seven patients received busulfan-containing myeloablative conditioning (MAC) regimens. Eleven patients underwent reduced intensity conditioning (RIC) regimens predominantly consisting of alemtuzumab, fludarabine, and melphalan. One patient received an intermediate-intensity regimen. Survival was poor in the MAC group, with only 1 patient surviving (14%). Most deaths were from transplantation-related toxicities, including venoocclusive disease and pulmonary hemorrhage. Of the 11 patients who received RIC, 6 are currently surviving at a median of 570 days after HCT (55%). Preparative regimen and HLH activity affected outcomes, and of RIC patients reported to be in remission from HLH, survival is 86% (P = .03). We conclude that MAC regimens should not be used for patients with XIAP deficiency. It is possible that the loss of XIAP and its antiapoptotic functions contributes to the high incidence of toxicities observed with MAC regimens. RIC regimens should be pursued with caution and, if possible, efforts should be made to ensure HLH remission before HCT in these patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Trastornos Linfoproliferativos/terapia , Acondicionamiento Pretrasplante/métodos , Adolescente , Niño , Preescolar , Terapia Combinada , Europa (Continente) , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hemorragia/etiología , Enfermedad Veno-Oclusiva Hepática/etiología , Humanos , Lactante , Japón , Pulmón/irrigación sanguínea , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/mortalidad , Mutación , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Inducción de Remisión , Estudios Retrospectivos , Análisis de Supervivencia , Tasa de Supervivencia , Trasplante Homólogo , Estados Unidos , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Adulto Joven
14.
Clin Immunol ; 149(3): 464-74, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24230999

RESUMEN

Defective V(D)J recombination and DNA double-strand break (DSB) repair severely impair the development of T-lymphocytes and B-lymphocytes. Most patients manifest a severe combined immunodeficiency during infancy. We report 2 siblings with combined immunodeficiency (CID) and immunodysregulation caused by compound heterozygous Artemis mutations, including an exon 1-3 deletion generating a null allele, and a missense change (p.T71P). Skin fibroblasts demonstrated normal DSB repair by gamma-H2AX analysis, supporting the predicted hypomorphic nature of the p.T71P allele. In addition to these two patients, 12 patients with Artemis-deficient CID were previously reported. All had significant morbidities including recurrent infections, autoimmunity, EBV-associated lymphoma, and carcinoma despite having hypomorphic mutants with residual Artemis expression, V(D)J recombination or DSB repair capacity. Nine patients underwent stem cell transplant and six survived, while four patients who did not receive transplant died. The progressive nature of immunodeficiency and genomic instability accounts for poor survival, and early HSCT should be considered.


Asunto(s)
Estudios de Asociación Genética , Mutación , Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/genética , Adulto , Secuencia de Aminoácidos , Preescolar , Proteínas de Unión al ADN , Endonucleasas , Femenino , Heterogeneidad Genética , Inestabilidad Genómica , Heterocigoto , Humanos , Lactante , Datos de Secuencia Molecular , Proteínas Nucleares/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Hermanos , Recombinación V(D)J/inmunología
15.
Cytotherapy ; 15(1): 109-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23260091

RESUMEN

BACKGROUND AIMS: Immunotherapy with allodepleted donor T cells improves immunity after T cell-depleted hematopoietic stem cell transplantation. We developed a methodology for selective depletion of alloreactive T cells after activation with host antigen-presenting cells by targeting T cells up-regulating CD25 and CD71. Combined depletion of these cells yields a pool of allodepleted donor T cells with antiviral properties with minimal capacity to cause graft-versus-host disease. METHODS: Mature dendritic cells were irradiated and used to stimulate donor peripheral blood mononuclear cells for 4 days. The co-culture was stained with anti-CD71-biotin followed by CliniMACS CD25 and Anti-Biotin Reagents (Miltenyi Biotec GmbH; Bergisch Gladbach, Germany) before depletion on the CliniMACS Plus (Miltenyi Biotec GmbH). Residual alloreactivity was tested by flow cytometry, a secondary mixed lymphocyte reaction and limiting dilution analysis, and specific anti-viral immunity with pentamer staining. The large-scale protocol was tested under current good manufacturing practice conditions in five donor-recipient pairs of human leukocyte antigen-matched volunteer donors. RESULTS: We developed a closed-system methodology using cell differentiation bags for cell culture and the COBE2991 Cell Processor (CaridianBCT, Lakewood, CO, USA). We also validated an anti-CD71-biotin generated for ex vivo clinical use. In five large-scale runs, the depleted fraction demonstrated excellent viability (99.9%), minimal residual expression of CD3/CD25 and CD3/CD71 (<0.2%) and passed tests for Mycoplasma, endotoxin, bacterial and fungal sterility. In secondary mixed lymphocyte reaction assays, the median response to host after allodepletion was 0%, whereas responses to third-party peripheral blood mononuclear cells were preserved (median, 105%; range 37%-350%). Limiting dilution analysis assays also demonstrated a reduction in response to host (median, -1.11 log) with preservation of third-party responses, and testing with human leukocyte antigen-restricted pentamers showed that populations of Epstein-Barr virus-specific and cytomegalovirus-specific CD8(+) T cells were retained after depletion. CONCLUSIONS: We optimized a protocol for the combined immunomagnetic depletion of alloreactive CD25/CD71 T cells under current good manufacturing practice conditions and tested the efficacy in five donor-recipient pairs.


Asunto(s)
Antígenos CD/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Dendríticas/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Depleción Linfocítica , Receptores de Transferrina/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucocitos Mononucleares/inmunología
16.
Pediatr Nephrol ; 28(10): 2053-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23743853

RESUMEN

BACKGROUND: Primary central nervous system (PCNS) post-transplant lymphoproliferative disorder (PTLD) is a rare complication of solid organ transplantation and is typically an Epstein-Barr virus (EBV)-induced B-cell CD20+ lymphoma. The modalities of treatment include reduction in immunosuppression, cranial radiotherapy (CRT), intravenous and intrathecal rituximab when CD20 is expressed on B-lymphocytes and PTLD cells, and chemotherapy. CASE-DIAGNOSIS/TREATMENT: We report the successful treatment of EBV-driven PCNS PTLD by reduction in immunosuppression (RI), CRT, and intravenous rituximab. Our patient was an 11-year-old boy with a living-related renal transplant for end-stage renal failure (ESRF) secondary to posterior urethral valves (PUV) and bilateral renal dysplasia (BRD) and on triple immunosuppression with prednisolone, tacrolimus, and azathioprine who had a rising EBV load, which was managed with reduction in tacrolimus dose, withdrawal of azathioprine, and introduction of mycophenolate mofetil (MMF). CONCLUSIONS: The patient presented 7 years post-transplant with a seizure and abnormal neurology secondary to polymorphous hyperplastic lesions in the brain, which responded to rituximab and CRT.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias del Sistema Nervioso Central/terapia , Quimioradioterapia , Irradiación Craneana , Infecciones por Virus de Epstein-Barr/virología , Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Linfoma de Células B/terapia , Antígenos CD20/metabolismo , Biopsia , Neoplasias del Sistema Nervioso Central/diagnóstico , Neoplasias del Sistema Nervioso Central/inmunología , Neoplasias del Sistema Nervioso Central/virología , Niño , Quimioterapia Combinada , Infecciones por Virus de Epstein-Barr/diagnóstico , Humanos , Inmunohistoquímica , Inmunosupresores/efectos adversos , Infusiones Intravenosas , Linfoma de Células B/diagnóstico , Linfoma de Células B/inmunología , Linfoma de Células B/virología , Imagen por Resonancia Magnética , Masculino , Rituximab , Convulsiones/virología , Factores de Tiempo , Resultado del Tratamiento
17.
Nat Med ; 29(7): 1700-1709, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37407840

RESUMEN

In the context of relapsed and refractory childhood pre-B cell acute lymphoblastic leukemia (R/R B-ALL), CD19-targeting chimeric antigen receptor (CAR)-T cells often induce durable remissions, which requires the persistence of CAR-T cells. In this study, we systematically analyzed CD19 CAR-T cells of 10 children with R/R B-ALL enrolled in the CARPALL trial via high-throughput single-cell gene expression and T cell receptor sequencing of infusion products and serial blood and bone marrow samples up to 5 years after infusion. We show that long-lived CAR-T cells developed a CD4/CD8 double-negative phenotype with an exhausted-like memory state and distinct transcriptional signature. This persistence signature was dominant among circulating CAR-T cells in all children with a long-lived treatment response for which sequencing data were sufficient (4/4, 100%). The signature was also present across T cell subsets and clonotypes, indicating that persisting CAR-T cells converge transcriptionally. This persistence signature was also detected in two adult patients with chronic lymphocytic leukemia with decade-long remissions who received a different CD19 CAR-T cell product. Examination of single T cell transcriptomes from a wide range of healthy and diseased tissues across children and adults indicated that the persistence signature may be specific to long-lived CAR-T cells. These findings raise the possibility that a universal transcriptional signature of clinically effective, persistent CD19 CAR-T cells exists.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Antígenos CD19/genética , Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfocitos T , Inducción de Remisión , Linfocitos T
18.
Blood Adv ; 7(9): 1725-1738, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36453632

RESUMEN

We recently described a low-affinity second-generation CD19 chimeric antigen receptor (CAR) CAT that showed enhanced expansion, cytotoxicity, and antitumor efficacy compared with the high-affinity (FMC63-based) CAR used in tisagenlecleucel, in preclinical models. Furthermore, CAT demonstrated an excellent toxicity profile, enhanced in vivo expansion, and long-term persistence in a phase 1 clinical study. To understand the molecular mechanisms behind these properties of CAT CAR T cells, we performed a systematic in vitro characterization of the transcriptomic (RNA sequencing) and protein (cytometry by time of flight) changes occurring in T cells expressing low-affinity vs high-affinity CD19 CARs following stimulation with CD19-expressing cells. Our results show that CAT CAR T cells exhibit enhanced activation to CD19 stimulation and a distinct transcriptomic and protein profile, with increased activation and cytokine polyfunctionality compared with FMC63 CAR T cells. We demonstrate that the enhanced functionality of low-affinity CAT CAR T cells is a consequence of an antigen-dependent priming induced by residual CD19-expressing B cells present in the manufacture.


Asunto(s)
Citocinas , Receptores Quiméricos de Antígenos , Citocinas/metabolismo , Inmunoterapia Adoptiva/métodos , Linfocitos T , Receptores Quiméricos de Antígenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD19
19.
Blood Adv ; 7(10): 2155-2165, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-36649566

RESUMEN

Stem cell transplant (SCT) outcomes in high-risk and relapsed/refractory (R/R) pediatric acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) have been historically poor. Cord blood (CB) allows T-cell replete CB transplant (TRCB), enabling enhanced graft-versus-leukemia. We consecutively collected data from 367 patients undergoing TRCB (112 patients) or other cell source (255 patients) SCT for pediatric AML/MDS in the United Kingdom and Ireland between January 2014 and December 2021. Data were collected about the patient's demographics, disease, and its treatment; including previous transplant, measurable residual disease (MRD) status at transplant, human leukocyte antigen-match, relapse, death, graft versus host disease (GvHD), and transplant-related mortality (TRM). Univariable and multivariable analyses were undertaken. There was a higher incidence of poor prognosis features in the TRCB cohort: 51.4% patients were MRD positive at transplant, 46.4% had refractory disease, and 21.4% had relapsed after a previous SCT, compared with 26.1%, 8.6%, and 5.1%, respectively, in the comparator group. Event free survival was 64.1% within the TRCB cohort, 50% in MRD-positive patients, and 79% in MRD-negative patients. To allow for the imbalance in baseline characteristics, a multivariable analysis was performed where the TRCB cohort had significantly improved event free survival, time to relapse, and reduced chronic GvHD, with some evidence of improved overall survival. The effect appeared similar regardless of the MRD status. CB transplant without serotherapy may be the optimal transplant option for children with myeloid malignancy.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Trastornos Mieloproliferativos , Humanos , Niño , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre/efectos adversos , Síndromes Mielodisplásicos/patología , Enfermedad Injerto contra Huésped/etiología , Leucemia Mieloide Aguda/patología , Recurrencia
20.
Br J Haematol ; 156(5): 656-66, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22224700

RESUMEN

Umbilical cord blood transplant (UCBT) is associated with impaired early immune reconstitution. This might be explained by a lower T-cell dose infused, the naivety of cord blood T-cells and the use of in vivo T-cell depletion. We studied the pattern of early immune reconstitution and the clinical outcome of children undergoing unrelated UCBT when in vivo T-cell depletion was omitted. Thirty children affected by malignancies (46%) or immunodeficiencies (54%) underwent an unrelated UCBT. Prospective assessment of immune reconstitution and clinical outcome was performed. We observed an unprecedented CD4(+) T-cell reconstitution, with a median cell count at 30 and 60 d post UCBT of 0.3 × 10(9) /l and 0.56 × 10(9) /l, respectively. Early T-cell expansion was thymic-independent, with a rapid shift from naïve to central memory phenotype and early regulatory T-cell recovery. Viral infections were frequent (63%) but resolved rapidly in most cases and virus-specific T-lymphocytes were detected within 2 months post-UCBT. Acute graft-versus-host disease (GvHD) was frequent (grade II = 34%, grade III-IV = 16%) but steroid responsive, and the incidence of chronic GvHD was low (14%). The omission of in vivo T-cell depletion promotes a unique thymic-independent CD4(+) T-cell reconstitution after unrelated UCBT in children. We postulate that this relates to the specific immunological and ontological qualities of fetal-derived lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Inmunidad Adaptativa , Linfocitos B/inmunología , Recuento de Linfocito CD4 , Niño , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Memoria Inmunológica , Inmunofenotipificación , Lactante , Células Asesinas Naturales/inmunología , Recuento de Linfocitos , Depleción Linfocítica , Masculino , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/prevención & control , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA