Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Oncol ; 39(29): 3261-3272, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34156898

RESUMEN

PURPOSE: Standard cytotoxic induction chemotherapy for acute myeloid leukemia (AML) results in prolonged neutropenia and risk of infection. Romyelocel-L is a universal, allogeneic myeloid progenitor cell product being studied to reduce infection during induction chemotherapy. PATIENTS AND METHODS: One hundred sixty-three patients with de novo AML (age ≥ 55 years) receiving induction chemotherapy were randomly assigned on day 0 (d0), of whom 120 were evaluable. Subjects received either romyelocel-L infusion on d9 with granulocyte colony-stimulating factor (G-CSF) starting daily d14 (treatment group) or G-CSF daily alone on d14 (control) until absolute neutrophil count recovery to 500/µL. End points included days in febrile episode, microbiologically defined infections, clinically diagnosed infection, and days in hospital. RESULTS: Mean days in febrile episode was shorter in the treatment arm from d15 through d28 (2.36 v 3.90; P = .02). Similarly, a trend toward decreased microbiologically defined infections and clinically diagnosed infection in the treatment arm was observed from d9 to d28 (35.6% v 47.5%; P = .09), reaching a statistically significant difference from d15 to d28 (6.8% v 27.9%; P = .002). Because of this, antibacterial or antifungal use for treatment of an infection was significantly less in the treatment group (d9-d28: 44.1% v 63.9%; P = .01). Significantly fewer patients in the treatment arm received empiric antifungals from d9 tod28 (42.4% v 63.9%; P = .02) and d15-d28 (42.4% v 62.3%; P = .02). Patients in the treatment arm also had 3.2 fewer hospital days compared with control (25.5 v 28.7; P = .001). Remission rates and days to absolute neutrophil count recovery were similar in the two groups. No patients in the romyelocel-L plus G-CSF group died because of infection compared with two patients in the control arm. No graft-versus-host disease was observed. CONCLUSION: Subjects receiving romyelocel-L showed a decreased incidence of infections, antimicrobial use, and hospitalization, suggesting that romyelocel-L may provide a new option to reduce infections in patients with AML undergoing induction therapy.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Células Progenitoras Mieloides/trasplante , Adulto , Anciano , Antifúngicos/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Quimioterapia de Inducción , Masculino , Persona de Mediana Edad , Neutrófilos/fisiología , Estudios Prospectivos
2.
Blood Adv ; 2(14): 1738-1749, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30037800

RESUMEN

The current standard of care for acute myeloid leukemia (AML) is largely ineffective with very high relapse rates and low survival rates, mostly due to the inability to eliminate a rare population of leukemic stem cells (LSCs) that initiate tumor growth and are resistant to standard chemotherapy. RNA-sequencing analysis on isolated LSCs confirmed C-type lectin domain family 12 member A (CLL1, also known as CLEC12A) to be highly expressed on LSCs but not on normal hematopoietic stem cells (HSCs) or other healthy organ tissues. Expression of CLL1 was consistent across different types of AML. We developed CLT030 (CLL1-ADC), an antibody-drug conjugate (ADC) based on a humanized anti-CLL1 antibody with 2 engineered cysteine residues linked covalently via a cleavable linker to a highly potent DNA-binding payload, thus resulting in a site-specific and homogenous ADC product. The ADC is designed to be stable in the bloodstream and to release its DNA-binding payload only after the ADC binds to CLL1-expressing tumor cells, is internalized, and the linker is cleaved in the lysosomal compartment. CLL1-ADC inhibits in vitro LSC colony formation and demonstrates robust in vivo efficacy in AML cell tumor models and tumor growth inhibition in the AML patient-derived xenograft model. CLL1-ADC demonstrated a reduced effect on differentiation of healthy normal human CD34+ cells to various lineages as observed in an in vitro colony formation assay and in an in vivo xenotransplantation model as compared with CD33-ADC. These results demonstrate that CLL1-ADC could be an effective ADC therapeutic for the treatment of AML.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunoconjugados/farmacología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda , Proteínas de Neoplasias/inmunología , Células Madre Neoplásicas , Receptores Mitogénicos/inmunología , Animales , Femenino , Células HL-60 , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Masculino , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Células Madre Neoplásicas/patología , Receptores Mitogénicos/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cloning Stem Cells ; 9(1): 51-62, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17386014

RESUMEN

The utilization of human hepatocytes for biomedical research, drug discovery, and treatment of liver diseases is hindered by the limited availability of donated livers and the variability of their derived hepatocytes. Human embryonic stem cells (hESCs) are pluripotent and provide a unique, unlimited resource for human hepatocytes. However, differentiation of hESCs to hepatocytes remains a challenge. We have developed a multistage procedure by which hESCs can be directly differentiated to hepatocyte-like cells without embryoid body formation and the requirement of sodium butyrate. The hESC-derived hepatocyte-like cells (HLCs) exhibited characteristic hepatocyte morphology, expressed hepatocyte markers, including alpha-fetoprotein, albumin, and hepatocyte nuclear factor 4alpha, and possessed hepatocyte-specific activities, such as p450 metabolism, albumin production, glycogen storage, and uptake and excretion of indocyanine green. Hepatocyte growth factor was found to play a positive role in promoting hepatocyte differentiation. Our differentiation system has shown that hESCs can be differentiated to hepatocyte-like cells capable of executing a range of hepatocyte functions. Therefore, it presents a proof-of-principle of potential applications of using the hESC-derived hepatocytes. Additionally, the hESC-derived HLCs provide a unique model to study the mechanisms involved in human hepatocyte differentiation and liver function.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Hepatocitos/fisiología , Células Madre Pluripotentes/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Hepatocitos/citología , Humanos , Hígado/citología , Hígado/fisiología , Células Madre Pluripotentes/citología
4.
Biotechnol Bioeng ; 91(6): 688-98, 2005 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-15971228

RESUMEN

Human embryonic stem cells (hESCs) can serve as an unlimited cell source for cellular transplantation and tissue engineering due to their prolonged proliferation capacity and their unique ability to differentiate into derivatives of all three-germ layers. In order to reliably and safely produce hESCs, use of reagents that are defined, qualified, and preferably derived from a non-animal source is desirable. Traditionally, mouse embryonic fibroblasts (MEFs) have been used as feeder cells to culture undifferentiated hESCs. We recently reported a scalable feeder-free culture system using medium conditioned by MEFs. The base and conditioned medium (CM) still contain unknown bovine and murine-derived components, respectively. In this study, we report the development of a hESC culture system that utilizes a commercially available serum-free medium (SFM) containing human sourced and recombinant proteins supplemented with recombinant growth factor(s) and does not require conditioning with feeder cells. In this system, which employs human laminin coated surface and high concentration of hbFGF, the hESCs maintained undifferentiated hESC morphology and had a twofold increase in expansion compared to hESCs grown in MEF-CM. The hESCs also expressed surface markers SSEA-4 and Tra-1-60 and maintained expression of hTERT, Oct4, and Cripto genes similar to cells cultured in MEF-CM. In addition, hESCs maintained in this culture system were able to differentiate in vitro and in vivo into cells of all three-germ layers. The cells maintained a normal karyotype after prolonged culture in SFM. In summary, this study demonstrates that the hESCs cultured in defined non-conditioned serum-free medium (NC-SFM) supplemented with growth factor(s) retain the characteristics and replicative potential of hESCs. The use of defined culture system with NC-SFM on human laminin simplifies scale-up and allows for reproducible generation of hESCs under defined and controlled conditions that would serve as a starting material for production of hESC derived cells for therapeutic use.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medio de Cultivo Libre de Suero/química , Embrión de Mamíferos/citología , Células Madre/citología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Medio de Cultivo Libre de Suero/farmacología , Relación Dosis-Respuesta a Droga , Factores de Crecimiento de Fibroblastos/farmacología , Sustancias de Crecimiento/administración & dosificación , Sustancias de Crecimiento/farmacología , Humanos , Laminina , Proteínas Recombinantes/farmacología , Células Madre/efectos de los fármacos
5.
Fertil Steril ; 83(5): 1517-29, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15866593

RESUMEN

OBJECTIVE: To derive new human embryonic stem cell (hESC) lines on pathogen-free human placental fibroblast feeders under serum-free conditions. Because the embryo develops in close contact with extraembryonic membranes, we hypothesized that placental mesenchyme might replicate the stem cell niche in situ. DESIGN: We isolated and characterized human placental fibroblast lines from individual donors and tested their ability to support growth of federally registered hESC lines. Moreover, we performed extensive pathogen testing to ensure their suitability as feeders for the derivation of therapy-grade hESCs. RESULT(S): Human placental fibroblasts were comparable or superior to mouse embryo fibroblasts as hESC feeders. We used these qualified placental fibroblasts to derive two new hESC lines in knockout Dulbecco's modified Eagle's medium with serum-free 20% knockout serum replacement. The cells, which had a normal karyotype, were grown for more than 25 passages, expressed markers of stemness including Oct-3/4, Tra 1-60, Tra 1-80, and SSEA-4, exhibited high telomerase activity, and differentiated in vitro and in vivo into cells derived from all three germ layers, confirming their pluripotency. Additionally, newly derived hESCs were adapted to growth on a human placental laminin substrate in a defined medium. CONCLUSION(S): To our knowledge, this is the first report of hESC derivation in the absence of serum on qualified pathogen-free human feeders.


Asunto(s)
Diferenciación Celular , Medio de Cultivo Libre de Suero/farmacología , Fibroblastos/citología , Placenta/citología , Técnicas de Cultivo de Tejidos/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Humanos , Placenta/efectos de los fármacos , Placenta/fisiología , Embarazo , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/fisiología
6.
Mil Med ; 167(2 Suppl): 78-81, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11873524

RESUMEN

Ex vivo expansion of a small volume of bone marrow offers an alternate approach to cellular support for repair of a hematopoietic system damaged by radiation exposure. Unpurified bone marrow cells cultured using frequent exchange of medium (perfusion) results in (1) the growth of an adherent layer that provides a supportive microenvironment for hematopoiesis and (2) the growth of hematopoietic stem and progenitor cells. Similar increases in stem and progenitor cells were also observed in cord blood cultures using frequent medium exchange methods. An automated clinical scale system, the AastromReplicell Cell Production System, has been developed to implement the biology of this ex vivo expansion process. Clinical use of ex vivo expanded bone marrow alone or in augmenting a low dose of mobilized peripheral blood stem cells has successfully reconstituted hematopoiesis. Ex vivo expanded cord blood cells combined with unmanipulated cord blood cells have also shown significant benefits in overall survival and engraftment in pediatric and adult patients, respectively. These results suggest that ex vivo expansion of hematopoietic cells provides a mechanism for generating cells capable of hematopoietic reconstitution.


Asunto(s)
Células de la Médula Ósea/fisiología , Sangre Fetal/citología , Células Madre Hematopoyéticas/fisiología , Expansión de Tejido , Adulto , Niño , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...