Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 9(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34266886

RESUMEN

Cell-based immunotherapies have had remarkable success in the clinic, specifically in the treatment of hematologic malignancies. However, these strategies have had limited efficacy in patients with solid tumors. To better understand the challenges involved, the National Cancer Institute (NCI) convened an initial workshop with immuno-oncology thought leaders in December 2018 and a follow-up workshop in December 2020. The goals of the NCI workshops on cell-based immunotherapy for solid tumors were to discuss the current state of the field of cell-based immunotherapy, obtain insights into critical knowledge gaps, and identify ways in which NCI could facilitate progress. At both meetings, subjects emphasized four main types of challenges in further developing cell-based immunotherapy for patients with solid tumors: scientific, technical, clinical, and regulatory. The scientific barriers include selecting appropriate targets, ensuring adequate trafficking of cell therapy products to tumor sites, overcoming the immunosuppressive tumor microenvironment, and identifying appropriate models for these investigations. While mouse models may provide some useful data, the majority of those that are commonly used are immunodeficient and unable to fully recapitulate the immune response in patients. There is therefore a need for enhanced support of small early-phase human clinical studies, preferably with adaptive trial designs, to provide proof of concept for novel cell therapy approaches. Furthermore, the requirements for manufacturing, shipping, and distributing cell-based therapies present technical challenges and regulatory questions, which many research institutions are not equipped to address. Overall, workshop subjects identified key areas where NCI support might help the research community in driving forward innovation and clinical utility: 1) provide focused research support on topics such as tumor target selection, immune cell fitness and persistence, cell trafficking, and the immunosuppressive tumor microenvironment; 2) support the rapid translation of preclinical findings into proof of concept clinical testing, harmonize clinical trial regimens, and facilitate early trial data sharing (including negative results); 3) expand manufacturing support for cell therapies, including vectors and reagents, and provide training programs for technical staff; and 4) develop and share standard operating procedures for cell handling and analytical assays, and work with the Food and Drug Administration to harmonize product characterization specifications.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Educación/normas , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Historia del Siglo XXI , Humanos , National Cancer Institute (U.S.) , Estados Unidos
2.
Nat Rev Drug Discov ; 15(9): 589-590, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27469032

RESUMEN

A global response to the chronic shortfall in antibiotic innovation is urgently needed to combat antimicrobial resistance. Here, we introduce CARB-X, a new global public-private partnership that will invest more than US$350 million in the next 5 years to accelerate the progression of a diverse portfolio of innovative antibacterial products into clinical trials.


Asunto(s)
Antibacterianos/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Farmacorresistencia Bacteriana , Asociación entre el Sector Público-Privado , Antibacterianos/uso terapéutico , Investigación Biomédica/economía , Investigación Biomédica/organización & administración , Ensayos Clínicos como Asunto , Humanos
3.
Gynecol Oncol ; 130(3): 518-24, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756180

RESUMEN

OBJECTIVE: The conditionally replicative adenovirus Ad5/3-Δ24 has a type-3 knob incorporated into the type-5 fiber that facilitates enhanced ovarian cancer infectivity. Preclinical studies have shown that Ad5/3-Δ24 achieves significant oncolysis and anti-tumor activity in ovarian cancer models. The purpose of this study was to evaluate in a phase I trial the feasibility and safety of intraperitoneal (IP) Ad5/3-Δ24 in recurrent ovarian cancer patients. METHODS: Eligible patients were treated with IP Ad5/3-Δ24 for 3 consecutive days in one of three dose cohorts ranging 1 × 10(10)-1 × 10(12)vp. Toxicity was assessed utilizing CTC grading and efficacy with RECIST. Ascites, serum, and other samples were obtained to evaluate gene transfer, generation of wildtype virus, viral shedding, and antibody response. RESULTS: Nine of 10 patients completed treatment per protocol. A total of 15 vector-related adverse events were experienced in 5 patients. These events included fever or chills, nausea, fatigue, and myalgia. All were grades 1-2 in nature, transient, and medically managed. Of the 8 treated patients evaluable for response, six patients had stable disease and 2 patients had progressive disease. Three patients had decreased CA-125 from pretreatment levels one month after treatment. Ancillary biologic studies indicated Ad5/3-Δ24 replication in patients in the higher dose cohorts. All patients experienced an anti-adenoviral neutralizing antibody effect. CONCLUSIONS: This study suggests the feasibility and safety of a serotype chimeric infectivity-enhanced CRAd, Ad5/3-Δ24, as a potential therapeutic option for recurrent ovarian cancer patients.


Asunto(s)
Adenoviridae , Anticuerpos Antivirales/sangre , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Ováricas/terapia , Adenoviridae/genética , Adenoviridae/fisiología , Anciano , Anciano de 80 o más Años , Ascitis/virología , Antígeno Ca-125/sangre , Escalofríos/virología , Progresión de la Enfermedad , Fatiga/virología , Femenino , Fiebre/virología , Expresión Génica , Humanos , Dosis Máxima Tolerada , Persona de Mediana Edad , Náusea/virología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/virología , Tomografía Computarizada por Rayos X , Replicación Viral , Esparcimiento de Virus
4.
Emerg Infect Dis ; 18(12): e2, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23171644

RESUMEN

The US Public Health Emergency Medical Countermeasures Enterprise convened subject matter experts at the 2010 HHS Burkholderia Workshop to develop consensus recommendations for postexposure prophylaxis against and treatment for Burkholderia pseudomallei and B. mallei infections, which cause melioidosis and glanders, respectively. Drugs recommended by consensus of the participants are ceftazidime or meropenem for initial intensive therapy, and trimethoprim/sulfamethoxazole or amoxicillin/clavulanic acid for eradication therapy. For postexposure prophylaxis, recommended drugs are trimethoprim/sulfamethoxazole or co-amoxiclav. To improve the timely diagnosis of melioidosis and glanders, further development and wide distribution of rapid diagnostic assays were also recommended. Standardized animal models and B. pseudomallei strains are needed for further development of therapeutic options. Training for laboratory technicians and physicians would facilitate better diagnosis and treatment options.


Asunto(s)
Antibacterianos/administración & dosificación , Burkholderia mallei/patogenicidad , Burkholderia pseudomallei/patogenicidad , Muermo/prevención & control , Melioidosis/prevención & control , Profilaxis Posexposición/métodos , Combinación Amoxicilina-Clavulanato de Potasio/administración & dosificación , Animales , Ceftazidima/administración & dosificación , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Muermo/diagnóstico , Muermo/tratamiento farmacológico , Humanos , Melioidosis/diagnóstico , Melioidosis/tratamiento farmacológico , Meropenem , Factores de Riesgo , Tienamicinas/administración & dosificación , Combinación Trimetoprim y Sulfametoxazol/administración & dosificación
5.
Clin Cancer Res ; 18(12): 3440-51, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22510347

RESUMEN

PURPOSE: Ad5.SSTR/TK.RGD is an infectivity-enhanced adenovirus expressing a therapeutic thymidine kinase suicide gene and a somatostatin receptor (SSTR) that allows for noninvasive gene transfer imaging. The purpose of this study was to identify the maximum tolerated dose (MTD), toxicities, clinical efficacy, and biologic effects of Ad5.SSTR/TK.RGD in patients with recurrent gynecologic cancer. EXPERIMENTAL DESIGN: Eligible patients were treated intraperitoneally for 3 days with 1 × 10(9) to 1 × 10(12) vp/dose of Ad5.SSTR/TK.RGD followed by intravenous ganciclovir for 14 days. Toxicity and clinical efficacy were assessed using Common Toxicity Criteria (CTC) Adverse Events grading and Response Evaluation Criteria in Solid Tumors (RECIST) criteria. Imaging using In-111 pentetreotide was obtained before and after treatment. Tissue samples were obtained to evaluate for gene transfer, generation of wild-type virus, viral shedding, and antibody response. RESULTS: Twelve patients were treated in three cohorts. The most common vector-related clinical toxicities were grade I/II constitutional or pain symptoms, experienced most often in patients treated at the highest dose. MTD was not identified. Five patients showed stable disease; all others experienced progressive disease. One patient with stable disease experienced complete resolution of disease and normalization of CA125 on further follow-up. Imaging detected increased In-111 pentetreotide retention in patients treated at the highest dose. Ancillary studies showed presence of Ad5.SSTR/TK.RGD virus and HSV1-tk expression in ascites samples collected at various time points in most patients treated within the higher dose cohorts. CONCLUSIONS: This study shows the safety, potential efficacy, and possible gene transfer imaging capacity of Ad5.SSTR/TK.RGD in patients with recurrent gynecologic cancer. Further development of this novel gene therapeutic appears to be warranted.


Asunto(s)
Adenoviridae/genética , Neoplasias Endometriales/terapia , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias Ováricas/terapia , Anciano , Antígeno Ca-125/sangre , Terapia Combinada , Neoplasias Endometriales/tratamiento farmacológico , Femenino , Ganciclovir/uso terapéutico , Vectores Genéticos , Humanos , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Somatostatina/genética , Somatostatina/biosíntesis , Replicación Viral
6.
Hum Gene Ther ; 22(7): 821-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21171861

RESUMEN

Conditionally replicative adenoviral (CRAd) virotherapy represents a promising therapeutic approach for cancer. We have demonstrated that a serotype chimeric adenoviral 5/3 fiber-knob modification achieves enhanced ovarian cancer infectivity, conditional replication, and oncolytic activity. This study evaluated the safety of intraperitoneal (IP) Ad5/3-Δ24 in advance of a phase I clinical trial in gynecologic cancers. Syrian hamster cohorts were treated with IP Ad5/3-Δ24 or control buffer for 3 consecutive days and euthanized on study days 8, 17, 57, and 89. Blood and tissue samples were harvested from each animal. For biodistribution studies, presence and quantitation of viral levels within samples were determined via quantitative polymerase chain reaction. For safety studies, animals were assessed for adverse vector-related tissue or laboratory effects. In the biodistribution study, low levels of Ad5/3-Δ24 DNA were noted outside of the abdominal cavity. Viral DNA levels in tissues obtained from the peritoneal cavity peaked at day 8 and declined thereafter. In the safety study, no specific histopathologic changes were attributable to virus administration. Hematologic findings noted in the 1 × 10(11) viral particles (vp)/dose group on Days 4 and/or 8 were indicative of an Ad5/3-Δ24-specific generalized inflammatory response; these findings resolved by day 56. The no observable adverse effect level was determined to be 1 × 10(10) vp/dose. This study elucidates the safety profile of IP administration of the serotype chimeric infectivity-enhanced CRAd, Ad5/3-Δ24, and provides guidance for a planned phase I trial for patients with recurrent gynecologic cancers.


Asunto(s)
Adenoviridae/genética , ADN Viral/genética , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/virología , Adenoviridae/fisiología , Animales , Anticuerpos Neutralizantes/sangre , Cricetinae , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética , Vectores Genéticos/farmacología , Inyecciones Intraperitoneales , Mesocricetus , Reacción en Cadena de la Polimerasa , Serotipificación , Distribución Tisular , Replicación Viral
7.
Clin Cancer Res ; 16(21): 5277-87, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20978148

RESUMEN

PURPOSE: To determine the maximum tolerated dose (MTD), toxicity spectrum, clinical activity, and biological effects of the tropism-modified, infectivity-enhanced conditionally replicative adenovirus (CRAd), Ad5-Δ24-Arg-Gly-Asp (RGD), in patients with malignant gynecologic diseases. EXPERIMENTAL DESIGN: Cohorts of eligible patients were treated daily for 3 days through an i.p. catheter. Vector doses ranged from 1 × 10(9) to 1 × 10(12) viral particles per day. Toxicity was evaluated using CTCv3.0. CA-125 and Response Evaluation Criteria in Solid Tumors (RECIST) criteria were used to determine clinical efficacy. Corollary biological studies included assessment of CRAd replication, wild-type virus generation, viral shedding, and neutralizing antibody response. RESULTS: Twenty-one patients were treated. Adverse clinical effects were limited to grade 1/2 fever, fatigue, or abdominal pain. No vector-related grade 3/4 toxicities were noted. No clinically significant laboratory abnormalities were noted. The maximum tolerated dose was not reached. Over a 1 month follow-up, 15 (71%) patients had stable disease and six (29%) had progressive disease. No partial or complete responses were noted. Seven patients had a decrease in CA-125; four had a >20% drop. RGD-specific PCR showed the presence of study vector in ascites of 16 patients. Seven revealed an increase in virus after day 3, suggesting replication of Ad5-Δ24-RGD. Minimal wild-type virus generation was detected. Viral shedding studies showed insignificant shedding in the serum, saliva, and urine. Anti-adenoviral neutralizing antibody effects were prevalent. CONCLUSIONS: This study, the first to evaluate an infectivity-enhanced CRAd in human cancer, shows the feasibility, safety, potential antitumor response, and biological activity of this approach in ovarian cancer. Further evaluation of infectivity enhanced virotherapy approaches for malignant gynecologic diseases is warranted.


Asunto(s)
Adenoviridae/genética , Adenoviridae/fisiología , Vacunas contra el Cáncer/uso terapéutico , Carcinoma/terapia , Neoplasias de los Genitales Femeninos/terapia , Tropismo Viral/genética , Replicación Viral/genética , Adenoviridae/patogenicidad , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Carcinoma/complicaciones , Carcinoma/virología , Femenino , Neoplasias de los Genitales Femeninos/complicaciones , Neoplasias de los Genitales Femeninos/virología , Humanos , Persona de Mediana Edad , Oligopéptidos/genética , Viroterapia Oncolítica/métodos , Organismos Modificados Genéticamente , Recurrencia , Células Tumorales Cultivadas , Vacunas de ADN/efectos adversos , Vacunas de ADN/genética , Vacunas de ADN/uso terapéutico
8.
Mol Pharmacol ; 70(5): 1488-93, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16896072

RESUMEN

Delivery of multiple exogenous genes into target cells is important for a broad range of gene therapy applications, including combined therapeutic gene expression and noninvasive imaging. Previous studies ( Mol Ther 4: 223-231, 2001 ) have described the adenoviral vector RGDTKSSTR with a double-expression cassette that encodes herpes simplex virus thymidine kinase (HSVtk) for molecular chemotherapy and human somatostatin receptor subtype-2 (hSSTR2) for indirect imaging. In this vector, both genes are inserted in place of the E1 region of the adenoviral genome and expressed independently from two cytomegalovirus (CMV) promoters. During production of clinical-grade RGDTKSSTR, we found that the CMV promoters and simian virus 40 (SV40) poly(A) regions located in both expression cassettes provoked homologous recombination and deletion of one of the cassettes. To resolve this problem, we designed a strategy for substituting the duplicate promoters and poly(A) regions. We placed the hSSTR2 gene in the new Ad5.SSTR/TK.RGD vector under the control of a CMV promoter with a bovine growth hormone poly(A) region, whereas the SV40 promoter, enhancer, and poly(A) signal controlled HSVtk expression. This use of different regulatory sequences allowed independent expression of both transgenes from a single adenoviral vector and circumvented the recombination problem. Reconstruction of the vector with a double-expression cassette enables its use in human clinical trials.


Asunto(s)
Adenoviridae/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Recombinación Genética/genética , Adenoviridae/fisiología , Secuencia de Bases , Células Cultivadas , Genoma Viral/genética , Humanos , Péptidos/química , Reproducibilidad de los Resultados , Eliminación de Secuencia , Homología de Secuencia , Transgenes
9.
Mol Biol Cell ; 13(12): 4484-96, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12475967

RESUMEN

The phosphorylated, activated cytoplasmic domains of the transforming growth factor-beta (TGFbeta) receptors were used as probes to screen an expression library that was prepared from a highly TGFbeta-responsive intestinal epithelial cell line. One of the TGFbeta receptor-interacting proteins isolated was identified to be the mammalian homologue of the LC7 family (mLC7) of dynein light chains (DLCs). This 11-kDa cytoplasmic protein interacts with the TGFbeta receptor complex intracellularly and is phosphorylated on serine residues after ligand-receptor engagement. Forced expression of mLC7-1 induces specific TGFbeta responses, including an activation of Jun N-terminal kinase (JNK), a phosphorylation of c-Jun, and an inhibition of cell growth. Furthermore, TGFbeta induces the recruitment of mLC7-1 to the intermediate chain of dynein. A kinase-deficient form of TGFbeta RII prevents both mLC7-1 phosphorylation and interaction with the dynein intermediate chain (DIC). This is the first demonstration of a link between cytoplasmic dynein and a natural growth inhibitory cytokine. Furthermore, our results suggest that TGFbeta pathway components may use a motor protein light chain as a receptor for the recruitment and transport of specific cargo along microtublules.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila , Proteínas Quinasas JNK Activadas por Mitógenos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Células COS , Proteínas Portadoras/química , Línea Celular , Citoplasma/metabolismo , Perros , Dineínas , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptores de Factores de Crecimiento Transformadores beta/química , Transducción de Señal , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...