Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Toxicol Sci ; 155(2): 400-408, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27815492

RESUMEN

Stathmin-1 (STMN1) is a microtubule-destabilizing protein which is overexpressed in cancer. Its overexpression is associated with poor prognosis and also serves as a predictive marker to taxane therapy. We have developed a proprietary bi-functional shRNA (bi-shRNA) platform to execute RNA interference (RNAi)-mediated gene silencing and a liposome-carrier complex to systemically deliver the pbi-shRNA plasmids. In vitro and in vivo testing demonstrated efficacy and specificity of pbi-shRNA plasmid in targeting STMN1 (Phadke, A. P., Jay, C. M., Wang, Z., Chen, S., Liu, S., Haddock, C., Kumar, P., Pappen, B. O., Rao, D. D., Templeton, N. S., et al. (2011). In vivo safety and antitumor efficacy of bifunctional small hairpin RNAs specific for the human Stathmin 1 oncoprotein. DNA Cell Biol. 30, 715-726.). Biodistribution and toxicology studies in bio-relevant Sprague Dawley rats with pbi-shRNA STMN1 lipoplex revealed that the plasmid DNA was delivered to a broad distribution of organs after a single subcutaneous injection. Specifically, plasmid was detected within the first week using QPCR (threshold 50 copies plasmid/1 µg genomic DNA) at the injection site, lung, spleen, blood, skin, ovary (limited), lymph nodes, and liver. It was not detected in the heart, testis or bone marrow. No plasmid was detected from any organ 30 days after injection. Treatment was well tolerated. Minimal inflammation/erythema was observed at the injection site. Circulating cytokine response was also examined by ELISA. The IL-6 levels were induced within 6 h then declined to the vehicle control level 72 h after the injection. TNFα induction was transiently observed 4 days after the DNA lipoplex treatment. In summary, the pbi-shRNA STMN1 lipoplex was well tolerated and displayed broad distribution after a single subcutaneous injection. The pre-clinical data has been filed to FDA and the pbi-shRNA STMN1 lipoplex is being investigated in a phase I clinical study.


Asunto(s)
Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Estatmina/antagonistas & inhibidores , Estatmina/genética , Animales , Femenino , Humanos , Inyecciones Subcutáneas , Interleucina-6/sangre , Masculino , Neoplasias/metabolismo , Neoplasias/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética , Ratas , Ratas Sprague-Dawley , Estatmina/administración & dosificación , Estatmina/metabolismo , Distribución Tisular
2.
Gynecol Oncol ; 143(3): 504-510, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27678295

RESUMEN

OBJECTIVES: The majority of women with Stage III/IV ovarian cancer who achieve clinical complete response with frontline standard of care will relapse within 2years. Vigil immunotherapy, a GMCSF/bi-shRNA furin DNA engineered autologous tumor cell (EATC) product, demonstrated safety and induction of circulating activated T-cells against autologous tumor in Phase I trial Senzer et al. (2012, 2013) . Our objectives for this study include evaluation of safety, immune response and recurrence free survival (RFS). METHODS: This is a Phase II crossover trial of Vigil (1.0×107 cells/intradermal injection/month for 4 to 12 doses) in Stage III/IV ovarian cancer patients achieving cCR (normal imaging, CA-125≤35units/ml, physical exam, and no symptoms suggestive of the presence of active disease) following primary surgical debulking and carboplatin/paclitaxel adjuvant or neoadjuvant chemotherapy. Patients received Vigil or standard of care during the maintenance period. RESULTS: Forty-two patients were entered into trial, 31 received Vigil and 11 received standard of care. No≥Grade 3 toxicity related to product was observed. A marked induction of circulating activated T-cell population was observed against individual, pre-processed autologous tumor in the Vigil arm as compared to pre-Vigil baseline using IFNγ ELISPOT response (30/31 negative ELISPOT pre Vigil to 31/31 positive ELISPOT post Vigil, median 134 spots). Moreover, in correlation with ELISPOT response, RFS from time of procurement was improved (mean 826days/median 604days in the Vigil arm from mean 481days/median 377days in the control arm, p=0.033). CONCLUSION: In conjunction with the demonstrated safety, the high rate of induction of T-cell activation and correlation with improvement in RFS justify further Phase II/III assessment of Vigil.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Carcinoma Endometrioide/tratamiento farmacológico , Procedimientos Quirúrgicos de Citorreducción , Neoplasias Quísticas, Mucinosas y Serosas/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Carboplatino/administración & dosificación , Carcinoma Endometrioide/patología , Estudios Cruzados , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante , Neoplasias Quísticas, Mucinosas y Serosas/patología , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación , Linfocitos T
3.
Mol Ther ; 24(8): 1412-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27166877

RESUMEN

The EWS/FLI1 fusion gene is well characterized as a driver of Ewing's sarcoma. Bi-shRNA EWS/FLI1 is a functional plasmid DNA construct that transcribes both siRNA and miRNA-like effectors each of which targets the identical type 1 translocation junction region of the EWS/FLI1 transcribed mRNA sequence. Previous preclinical and clinical studies confirm the safety of this RNA interference platform technology and consistently demonstrate designated mRNA and protein target knockdown at greater than 90% efficiency. We initiated development of pbi-shRNA EWS/FLI1 lipoplex (LPX) for the treatment of type 1 Ewing's sarcoma. Clinical-grade plasmid was manufactured and both sequence and activity verified. Target protein and RNA knockdown of 85-92% was demonstrated in vitro in type 1 human Ewing's sarcoma tumor cell lines with the optimal bi-shRNA EWS/FLI1 plasmid. This functional plasmid was placed in a clinically tested, liposomal (LP) delivery vehicle followed by in vivo verification of activity. Type 1 Ewing's sarcoma xenograft modeling confirmed dose related safety and tumor response to pbi-shRNA EWS/FLI1 LPX. Toxicology studies in mini-pigs with doses comparable to the demonstrated in vivo efficacy dose resulted in transient fever, occasional limited hypertension at low- and high-dose assessment and transient liver enzyme elevation at high dose. These results provide the justification to initiate clinical testing.


Asunto(s)
Liposomas , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , ARN Interferente Pequeño/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Animales , Sitios de Unión , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Técnicas de Silenciamiento del Gen , Orden Génico , Terapia Genética , Humanos , Mediadores de Inflamación , Masculino , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/química , Plásmidos/administración & dosificación , Plásmidos/genética , Proteína Proto-Oncogénica c-fli-1/administración & dosificación , Proteína Proto-Oncogénica c-fli-1/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Proteína EWS de Unión a ARN/administración & dosificación , Proteína EWS de Unión a ARN/química , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/patología , Sarcoma de Ewing/terapia , Reparación del Gen Blanco , Transfección , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Ther ; 23(6): 1103-1109, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25917459

RESUMEN

We report on 12 consecutive patients with advanced/metastatic Ewing's sarcoma who were treated as a separate cohort of a phase 1 trial of FANG autologous immunotherapy (1 × 10(6)-2.5 × 10(7) cells/intradermal injection each month for minimum 4 months). Safety and clinical response were monitored. Patient immune response to unmodified autologous tumor cells was assessed by gamma interferon-enzyme-linked immunospot (γIFN-ELISPOT) assay using peripheral blood mononuclear cells from baseline (pretreatment) and multiple postvaccination time points. None of the 12 patients (47 vaccinations) developed grade 2/3/4 drug-related toxicity. Median product release granulocyte-macrophage colony-stimulating factor expression was 1,941 pg/10(6) cells, and TGFß1and TGFß2 knockdown were 99 and 100%, respectively. Eight patients were assessed for ELISPOT response to autologous tumor cells at baseline and all (100%) were negative. In contrast, follow-up ELISPOT response at month 1 or month 4 (one patient) after FANG was positive in all eight patients. One patient achieved a partial tumor response (38% tumor reduction, RECIST 1.1). The Kaplan-Meier estimated survival of these 12 patients at 1 year was 75%. In this phase 1 study in patients with Ewing's sarcoma, FANG immunotherapy was well tolerated, elicited a tumor-specific systemic immune response in all patients, and was associated with favorable 1-year survival. Further clinical testing is indicated.


Asunto(s)
Inmunoterapia/métodos , Sarcoma de Ewing/terapia , Adolescente , Adulto , Femenino , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/terapia , Proyectos Piloto , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Resultado del Tratamiento , Adulto Joven
5.
Mol Ther ; 23(6): 1123-1130, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25619726

RESUMEN

Stathmin1 (STMN1) is a microtubule modulator that is expressed in multiple cancers and correlates with poor survival. We previously demonstrated in vivo safety of bifunctional (bi) shRNA STMN1 bilamellar invaginated vesicle (BIV) and that systemic delivery correlated with antitumor activity. Patients with superficial advanced refractory cancer with no other standard options were entered into trial. Study design involved dose escalation (four patients/cohort) using a modified Fibonacci schema starting at 0.7 mg DNA administered via single intratumoral injection. Biopsy at baseline, 24/48 hours and resection 8 days after injection provided tissue for determination of cleavage product using next-generation sequencing (NGS) and reverse transcription quantitative polymerase chain reaction (RT-qPCR), 5' RLM rapid amplification of cDNA ends (RACE) assay. Serum pharmacokinetics of circulating plasmid was done. Twelve patients were entered into three dose levels (0.7, 1.4, 7.0 mg DNA). No ≥ grade 3 toxic effects to drug were observed. Maximum circulating plasmid was detected at 30 seconds with less than 10% detectable in all subjects at 24 hours. No toxic effects were observed. Predicted cleavage product was detected by both NGS (n = 7/7 patients analyzed, cohorts 1, 2) and RLM RACE (n = 1/1 patients analyzed cohort 3). In conclusion, bi-shRNA STMN1 BIV is well tolerated and detection of mRNA target sequence-specific cleavage product confirmed bi-shRNA BIV mechanism of action.


Asunto(s)
Neoplasias/terapia , ARN Interferente Pequeño/metabolismo , Estatmina/genética , Moduladores de Tubulina/farmacología , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inyecciones Intralesiones , Masculino , Persona de Mediana Edad , Neoplasias/genética , Plásmidos/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Estatmina/farmacocinética , Pruebas de Toxicidad
6.
Oncology ; 87(1): 21-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24968881

RESUMEN

Therapies for advanced hepatocellular carcinoma (HCC) are limited. We carried out a phase I trial of a novel autologous whole-cell tumor cell immunotherapy (FANG™), which incorporates a dual granulocyte macrophage colony-stimulating factor (GM-CSF) expressive/bifunctional small hairpin RNA interference (bi-shRNAi) vector. The bi-shRNAi DNA targets furin, which is a proconvertase of transforming growth factors beta (TGFß) 1 and 2. Safety, mechanism, immunoeffectiveness, and suggested benefit were previously shown [Senzer et al.: Mol Ther 2012;20:679-689; Senzer et al.: J Vaccines Vaccin 2013;4:209]. We now provide further follow-up of a subset of 8 HCC patients. FANG manufacturing was successful in 7 of 8 attempts (one failure due to insufficient cell yield). Median GM-CSF expression was 144 pg/10(6) cells, TGFß1 knockdown was 100%, and TGFß2 knockdown was 93% of the vector-transported cells. Five patients were vaccinated (1 or 2.5×10(7) cells/intradermal injection, 6-11 vaccinations). No FANG toxicity was observed. Three of these patients demonstrated evidence of an immune response to the autologous tumor cell sample. Long-term follow-up demonstrated survival of 319, 729, 784, 931+, and 1,043+ days of the FANG-treated patients. In conclusion, evidence supports further assessment of the FANG immunotherapy in HCC.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Carcinoma Hepatocelular/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias Hepáticas/terapia , ARN Interferente Pequeño/genética , Vacunación , Adulto , Anciano , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Inyecciones Intradérmicas , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Trasplante Autólogo , Resultado del Tratamiento
7.
Methods Mol Biol ; 942: 259-78, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23027056

RESUMEN

The discovery of RNA interference (RNAi) engendered great excitement and raised expectations regarding its potential applications in biomedical research and clinical usage. Over the ensuing years, expanded understanding of RNAi and preliminary results from early clinical trials tempered enthusiasm with realistic appraisal resulting in cautious optimism and a better understanding of necessary research and clinical directions. As a result, data from more recent trials are beginning to show encouraging positive clinical outcomes. The capability of delivering a pharmacologically effective dose to the target site while avoiding adverse host reactions still remains a challenge although the delivery technology continues to improve. We have developed a novel vector-driven bifunctional short hairpin RNA (bi-shRNA) technology that harnesses both cleavage-dependent and cleavage-independent RISC loading pathways to enhance knockdown potency. Consequent advantages provided by the bi-shRNA include a lower effective systemic dose than comparator siRNA/shRNA to minimize the potential for off-target side effects, due to its ability to induce both a rapid (inhibition of protein translation) and delayed (mRNA cleavage and degradation) targeting effect depending on protein and mRNA kinetics, and a longer duration of effectiveness for clinical applications. Here, we provide an overview of key molecular methods for the design, construction, quality control, and application of bi-shRNA that we believe will be useful for others interested in utilizing this technology.


Asunto(s)
Ingeniería Genética/métodos , ARN Interferente Pequeño/genética , Secuencia de Bases , Colesterol/metabolismo , ADN/genética , ADN/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Secuencias Invertidas Repetidas , Compuestos de Amonio Cuaternario/metabolismo , División del ARN , Interferencia de ARN , Transfección
8.
Mol Ther ; 20(3): 679-86, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22186789

RESUMEN

We performed a phase I trial of FANG vaccine, an autologous tumor-based product incorporating a plasmid encoding granulocyte-macrophage colony-stimulating factor (GMCSF) and a novel bifunctional short hairpin RNAi (bi-shRNAi) targeting furin convertase, thereby downregulating endogenous immunosuppressive transforming growth factors (TGF) ß1 and ß2. Patients with advanced cancer received up to 12 monthly intradermal injections of FANG vaccine (1 × 10(7) or 2.5 × 10(7) cells/ml injection). GMCSF, TGFß1, TGFß2, and furin proteins were quantified by enzyme-linked immunosorbent assay (ELISA). Safety and response were monitored. Vaccine manufacturing was successful in 42 of 46 patients of whom 27 received ≥1 vaccine. There were no treatment-related serious adverse events. Most common grade 1, 2 adverse events included local induration (n = 14) and local erythema (n = 11) at injection site. Post-transfection mean product expression GMCSF increased from 7.3 to 1,108 pg/10(6) cells/ml. Mean TGFß1 and ß2 effective target knockdown was 93.5 and 92.5% from baseline, respectively. Positive enzyme-linked immunospot (ELISPOT) response at month 4 was demonstrated in 9 of 18 patients serially assessed and correlated with survival duration from time of treatment (P = 0.025). Neither dose-adverse event nor dose-response relationship was noted. In conclusion, FANG vaccine was safe and elicited an immune response correlating with prolonged survival. Phase II assessment is justified.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Furina/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/terapia , ARN Interferente Pequeño/uso terapéutico , Adulto , Anciano , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Femenino , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interferón gamma/biosíntesis , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/inmunología , Neoplasias/mortalidad , Neoplasias/patología , Análisis de Supervivencia , Transgenes , Resultado del Tratamiento
9.
DNA Cell Biol ; 30(9): 715-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21612405

RESUMEN

Bifunctional small hairpin RNAs (bi-shRNAs) are functional miRNA/siRNA composites that are optimized for posttranscriptional gene silencing through concurrent mRNA cleavage-dependent and -independent mechanisms (Rao et al., 2010 ). We have generated a novel bi-shRNA using the miR30 scaffold that is highly effective for knockdown of human stathmin (STMN1) mRNA. STMN1 overexpression well documented in human solid cancers correlates with their poor prognosis. Transfection with the bi-shSTMN1-encoding expression plasmid (pbi-shSTMN1) markedly reduced CCL-247 human colorectal cancer and SK-Mel-28 melanoma cell growth in vitro (Rao et al., 2010 ). We now examine in vivo the antitumor efficacy of this RNA interference-based approach with human tumor xenografted athymic mice. A single intratumoral (IT) injection of pbi-shSTMN1 (8 µg) reduced CCL-247 tumor xenograft growth by 44% at 7 days when delivered as a 1,2-dioleoyl-3-trimethyl-ammoniopropane:cholesterol liposomal complex. Extended growth reductions (57% at day 15; p < 0.05) were achieved with three daily treatments of the same construct. STMN1 protein reduction was confirmed by immunoblot analysis. IT treatments with pbi-shSTMN1 similarly inhibited the growth of tumorgrafts derived from low-passage primary melanoma (≥70% reduction for 2 weeks) and abrogated osteosarcoma tumorgraft growth, with the mature bi-shRNA effector molecule detectable for up to 16 days after last injection. Antitumor efficacy was evident for up to 25 days posttreatment in the melanoma tumorgraft model. The maximum tolerated dose by IT injection of >92 µg (Human equivalent dose [HED] of >0.3 mg/kg) in CCL-247 tumor xenograft-bearing athymic mice was ∼10-fold higher than the extrapolated IC(50) of 9 µg (HED of 0.03 mg/kg). Healthy, immunocompetent rats were used as biorelevant models for systemic safety assessments. The observed maximum tolerated dose of <100 µg for intravenously injected pbi-shSTMN1 (mouse equivalent of <26.5 µg; HED of <0.09 mg/kg) confirmed systemic safety of the therapeutic dose, hence supporting early-phase assessments of clinical safety and preliminary efficacy.


Asunto(s)
Neoplasias Colorrectales/terapia , Técnicas de Silenciamiento del Gen/métodos , Terapia Genética/métodos , Melanoma/terapia , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Estatmina/metabolismo , Animales , Cartilla de ADN/genética , Femenino , Humanos , Immunoblotting , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Desnudos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Clin Cancer Res ; 17(1): 183-92, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21208907

RESUMEN

PURPOSE: On the basis of the hypothesis that the combined expression of immunostimulatory granulocyte macrophage colony stimulating factor (GM-CSF) and antitumor suppressor TGF-ß2 antisense (AS) transgenes can break tolerance and stimulate immune responses to cancer-associated antigens, we constructed an expression plasmid [the tumor-associated glycoprotein (TAG) plasmid] that coexpresses GM-CSF and TGF-ß2 AS nucleotide sequences and which was incorporated into an autologous whole-cell vaccine. EXPERIMENTAL DESIGN: Patients undergoing resection were enrolled. Freshly harvested autologous tumor cells were mechanically and enzymatically disaggregated, then electroporated with the TAG vector. The resulting vaccine was irradiated, then aliquoted and cryopreserved until the time of injection. Patients received a minimum of 5 to a maximum of 12 monthly intradermal injections. Immune function was monitored at baseline and at months 3 and 6. RESULTS: Vaccine manufacturing efficiency was 84% (32/38). Twenty-three patients received at least 1 vaccination. There were no grade 3 or 4 toxicities, and grade 1 and 2 events were local in nature. Seventeen of 21 patients had stable disease (SD) at month 2 or later as their best response, and 1 patient with stage IVa malignant melanoma achieved a complete response (CR) following 11 vaccinations and remains without evidence of disease 2 years following initiation of therapy. Six of 13 patients displayed a positive enzyme-linked immunospot (ELISPOT) response to autologous TAG vaccine at week 12 including 3 patients with prolonged SD or CR. The 3 other patients survived through week 24, as compared with none of the 7 ELISPOT-negative patients. CONCLUSIONS: On the basis of safety and clinical and immunologic results, further evaluation of bifunctional vaccines is warranted.


Asunto(s)
Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/terapia , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Factor de Crecimiento Transformador beta2/genética , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/inmunología , Trasplante Autólogo
11.
J Clin Oncol ; 24(29): 4721-30, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16966690

RESUMEN

PURPOSE: Belagenpumatucel-L is a nonviral gene-based allogeneic tumor cell vaccine that demonstrates enhancement of tumor antigen recognition as a result of transforming growth factor beta-2 inhibition. PATIENTS AND METHODS: We performed a randomized, dose-variable, phase II trial involving stages II, IIIA, IIIB, and IV non-small-cell lung cancer patients. Each patient received one of three doses (1.25, 2.5, or 5.0 x 10(7) cells/injection) of belagenpumatucel-L on a monthly or every other month schedule to a maximum of 16 injections. Immune function, safety, and anticancer activity were monitored. RESULTS: Seventy-five patients (two stage II, 12 stage IIIA, 15 stage IIIB, and 46 stage IV patients) received a total of 550 vaccinations. No significant adverse events were observed. A dose-related survival difference was demonstrated in patients who received > or = 2.5 x 10(7) cells/injection (P = .0069). Focusing on the 61 late-stage (IIIB and IV) assessable patients, a 15% partial response rate was achieved. The estimated probabilities of surviving 1 and 2 years were 68% and 52%, respectively for the higher dose groups combined and 39% and 20%, respectively, for the low-dose group. Immune function was explored in the 61 advanced-stage (IIIB and IV) patients. Increased cytokine production (at week 12 compared with patients with progressive disease) was observed among clinical responders (interferon gamma, P = .006; interleukin [IL] -6, P = .004; IL-4, P = .007), who also displayed an elevated antibody-mediated response to vaccine HLAs (P = .014). Furthermore, positive enzyme-linked immunospot reactions to belagenpumatucel-L showed a correlation trend (P = .086) with clinical responsiveness in patients achieving stable disease or better. CONCLUSION: Belagenpumatucel-L is well tolerated, and the survival advantage justifies further phase III evaluation.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Oligonucleótidos Antisentido/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Formación de Anticuerpos/efectos de los fármacos , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Oligonucleótidos Antisentido/efectos adversos , Oligonucleótidos Antisentido/inmunología , Análisis de Supervivencia , Factor de Crecimiento Transformador beta2 , Resultado del Tratamiento
12.
Clin Lymphoma ; 5(4): 285-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15794867

RESUMEN

Immunoglobulin (Ig) M myeloma is a distinct entity with features of multiple myeloma (MM) and Waldenstrom's macroglobulinemia (WM). The malignant cells in IgM myeloma have a distinctive chromosomal translocation that differentiates them from WM. These cells are postgerminal-center in origin with isotype-switch transcripts. They appear to be arrested at a point of maturation between that of WM and MM. Preliminary data indicate that a pattern of osteoclast-activating factor and osteoprotegerin expression similar to that observed in classic MM is present in IgM myeloma. Additional studies on patients with this rare tumor may provide further insight into the pathogenesis of bone disease in plasma cell dyscrasias.


Asunto(s)
Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/inmunología , Diferenciación Celular , Citocinas/biosíntesis , Glicoproteínas/biosíntesis , Humanos , Linfocinas/biosíntesis , Masculino , Persona de Mediana Edad , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores del Factor de Necrosis Tumoral , Translocación Genética
13.
Microvasc Res ; 67(2): 168-81, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15020208

RESUMEN

CAP37, a neutrophil-derived protein, originally identified for its antimicrobial activity is now known to have strong immunoregulatory effects on host cells. Recently, we described its expression and localization within the vascular endothelium associated with atherosclerotic plaques. Since CAP37 is a potent activator of endothelial cells and monocytes, two of the key cellular components of the atherosclerotic plaque, this study was undertaken to determine whether CAP37 had functional effects on smooth muscle cells another important cellular participant in atherosclerosis. Sections from atherosclerotic lesions were stained for the presence of CAP37 and smooth muscle cell alpha actin. The effect of CAP37 on aorta smooth muscle cell migration and proliferation was investigated and the upregulation of adhesion molecules was determined. Immunocytochemistry indicated that CAP37 was present in a subset of smooth muscle cells within atherosclerotic lesions, but was absent in normal vessels. Flow cytometry using double labeling for the proliferation marker Ki-67 and CAP37 demonstrates that CAP37 is mainly expressed in proliferating smooth muscle cells. We show that CAP37 supports migration and proliferation of smooth muscle cells in vitro. Furthermore, CAP37-treated smooth muscle cells expressed higher levels of the cell adhesion molecule ICAM-1 when compared with untreated cells. We suggest that due to its localization to atherosclerotic plaques and its ability to modulate smooth muscle cells, CAP37 may play a role in the progression of this disease.


Asunto(s)
Proteínas Sanguíneas/fisiología , Proteínas Portadoras/fisiología , Mediadores de Inflamación/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Adulto , Anciano , Péptidos Catiónicos Antimicrobianos , Aorta/citología , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/farmacología , Arterias Carótidas/citología , Estenosis Carotídea/patología , Proteínas Portadoras/análisis , Proteínas Portadoras/farmacología , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Quimiotaxis/efectos de los fármacos , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Antígeno Ki-67/análisis , Persona de Mediana Edad , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/fisiología , Isoformas de Proteínas/análisis , Proteínas Recombinantes/farmacología
14.
Microvasc Res ; 66(1): 38-48, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12826073

RESUMEN

Cationic antimicrobial protein of molecular weight 37 kDa (CAP37) is a multifunctional inflammatory mediator that was originally isolated from human neutrophils and described to possess bactericidal and monocyte-activating functions. More recently its expression in endothelial and epithelial cells in response to inflammatory mediators and its ability to activate endothelial cells and alter permeability has been demonstrated. We hypothesize that CAP37 facilitates the process of transendothelial migration not only because of its potential to act as a chemoattractant but also through its ability to promote leukocyte adhesion to the endothelium by modulating adhesion molecule expression on the endothelium. Here we describe its ability to mediate neutrophil and monocyte adherence to endothelial monolayers in vitro. Using reverse transcriptase-polymerase chain reaction and flow cytometry, we demonstrate its ability to upregulate the adhesion molecules, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin in human umbilical vein and lung microvessel endothelial cells. The identity and kinetics of upregulation of the specific adhesion molecule was dependent on the endothelial cell type, suggesting that adhesion molecules on endothelial cells from different vascular beds are differentially regulated by CAP37. The cell-specific kinetics of adhesion molecule upregulation by CAP37 may influence selective leukocyte migration in certain inflammatory situations.


Asunto(s)
Proteínas Sanguíneas/fisiología , Proteínas Portadoras/fisiología , Células Endoteliales/citología , Leucocitos/metabolismo , Péptidos Catiónicos Antimicrobianos , Cationes , Adhesión Celular , Movimiento Celular , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Selectina E/metabolismo , Endotelio Vascular/metabolismo , Células Epiteliales/citología , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Inflamación , Cinética , Leucocitos/citología , Microcirculación , Monocitos/metabolismo , Neutrófilos/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Venas Umbilicales/citología , Regulación hacia Arriba
15.
Glia ; 41(1): 64-72, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12465046

RESUMEN

Recent evidence suggests that inflammation and immune function in the central nervous system (CNS) may play a considerable role in the progression of many neurodegenerative diseases. It is known that microglia, the CNS equivalent of peripheral blood monocytes, may be instrumental in causing neurotoxicity. However, the mediator(s) that activates microglia to produce toxic substances that orchestrate cell death has yet to be elucidated. We have identified a novel inflammatory molecule, cationic antimicrobial protein of molecular weight 37 kDa (CAP37), to the brains of patients dying from Alzheimer's disease. CAP37 is known to be a potent activator and regulator of monocyte function in the systemic circulation. We hypothesize that CAP37, a mediator previously shown to recruit and activate monocytes in the systemic circulation, may also play a role in CNS inflammation by modulating microglial function. Here we demonstrate that CAP37 is a chemoattractant for microglia and that CAP37-treated microglia express class II major histocompatibility antigens and produce proinflammatory cytokines and chemokines. We conclude that CAP37 has the ability to activate microglial cells and suggest that it has the potential to serve as a neuroinflammatory molecule.


Asunto(s)
Proteínas Sanguíneas/fisiología , Proteínas Portadoras/fisiología , Mediadores de Inflamación/fisiología , Microglía/efectos de los fármacos , Microglía/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos , Proteínas Sanguíneas/farmacología , Proteínas Portadoras/farmacología , Línea Celular , Quimiocinas/biosíntesis , Quimiotaxis/efectos de los fármacos , Citocinas/biosíntesis , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Ratones , Microglía/citología , Fagocitosis/efectos de los fármacos
16.
Invest Ophthalmol Vis Sci ; 43(5): 1414-21, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11980855

RESUMEN

PURPOSE: CAP37 is a polymorphonuclear neutrophil (PMN)-derived inflammatory protein with potent antibiotic and chemotactic activity. To further investigate the biological significance of CAP37 in infection and inflammation, a well-characterized in vivo rabbit model of bacterial keratitis was selected to study its contribution to host defenses. METHODS: One hundred colony-forming units of log phase Staphylococcus aureus was injected intrastromally. Eyes were enucleated at 5 to 25 hours after infection and CAP37 detected by immunohistochemistry. To identify the mechanism of CAP37 upregulation in corneal epithelium, in vitro studies using immortalized human corneal epithelial cells (HCECs) were undertaken to determine whether proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta), induce CAP37. Because adhesion of leukocytes is important in leukocyte-epithelium interactions, the effect of CAP37 on expression of intercellular adhesion molecule (ICAM)-1 on HCECs was determined by flow cytometry. RESULTS: Strong staining for CAP37 was demonstrated in the corneal epithelium, stromal fibroblasts, ciliary epithelium, related limbus, ciliary vascular endothelium, and bulbar conjunctiva in rabbits injected with S. aureus. The most dramatic expression of CAP37 aside from that in the PMNs occurred in the corneal epithelium. The in vitro studies suggest that CAP37 induction is regulated by TNF-alpha and IL-1beta. In addition, ICAM-1 expression on HCECs was increased in response to CAP37. Molecular cloning of corneal epithelial CAP37 indicated strong sequence identity with an extensive region of PMN-CAP37. CONCLUSIONS: The findings in this study describe the extraneutrophilic expression of CAP37 in response to infection and suggest a role for CAP37 in host defense against infection in the eye.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Proteínas Portadoras/metabolismo , Epitelio Corneal/metabolismo , Infecciones Bacterianas del Ojo/metabolismo , Proteínas del Ojo/metabolismo , Queratitis/metabolismo , Infecciones Estafilocócicas/metabolismo , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Células Cultivadas , Clonación Molecular , Epitelio Corneal/efectos de los fármacos , Infecciones Bacterianas del Ojo/microbiología , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Fibroblastos/metabolismo , Citometría de Flujo , Técnicas para Inmunoenzimas , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1/farmacología , Queratitis/microbiología , Datos de Secuencia Molecular , Proteínas Quimioatrayentes de Monocitos/genética , Proteínas Quimioatrayentes de Monocitos/metabolismo , Proteínas Quimioatrayentes de Monocitos/farmacología , Conejos , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estafilocócicas/microbiología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
17.
Am J Pathol ; 160(3): 841-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11891183

RESUMEN

Cationic antimicrobial protein of 37 kd (CAP37), originally isolated from human neutrophils, is an important multifunctional inflammatory mediator. Here we describe its localization within the vascular endothelium associated with atherosclerotic plaques. Evidence from in vitro immunocytochemical, Northern blot, and reverse transcriptase-polymerase chain reaction analysis indicates that CAP37 is induced in endothelial cells in response to inflammatory mediators. Endothelial-derived CAP37 shows sequence identity with an extensive region of neutrophil-derived CAP37. This is the first demonstration of endogenous endothelial CAP37, confirmed by sequence analysis. We suggest that, because of its induction and location in the endothelium and its known monocyte- and endothelial-activating capabilities, CAP37 has potential to modulate monocyte/endothelial dynamics at the vessel wall in inflammation.


Asunto(s)
Arteriosclerosis/metabolismo , Proteínas Sanguíneas/biosíntesis , Proteínas Portadoras/biosíntesis , Endotelio Vascular/metabolismo , Mediadores de Inflamación/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos , Arteriosclerosis/inmunología , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Endotelio Vascular/inmunología , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Mediadores de Inflamación/inmunología , Reacción en Cadena de la Polimerasa , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...