Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Ann Oncol ; 33(10): 1052-1060, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35764271

RESUMEN

BACKGROUND: In the phase II multicohort CheckMate 142 study, nivolumab plus low-dose (1 mg/kg) ipilimumab provided robust and durable clinical benefit with a manageable safety profile in previously treated patients with microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC) at 13.4- and 25.4-month median follow-up (Overman MJ, Lonardi S, Wong KYM et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018;36:773-779. Overman MJ, Lonardi S, Wong KYM, et al. Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer: long-term follow-up. J Clin Oncol. 2019;37:635). Here, we present results from the 4-year follow-up of these patients. PATIENTS AND METHODS: Patients received nivolumab (3 mg/kg) plus low-dose (1 mg/kg) ipilimumab every 3 weeks (four doses) followed by nivolumab (3 mg/kg) every 2 weeks until disease progression. Primary endpoint was investigator-assessed objective response rate (ORR; as per RECIST version 1.1). RESULTS: A total of 119 patients were treated; 76% had ≥2 prior lines of therapy. Median follow-up was 50.9 months (range 46.9-62.7 months). Median duration of therapy was 24.9 months [95% confidence interval (CI) 15.8-33.2 months]. Investigator-assessed ORR increased from 55% (95% CI 45% to 64%) at 13.4 months to 65% (95% CI 55% to 73%) at 50.9 months with a disease control rate of 81% (95% CI 72% to 87%). The complete response rate increased from 3% at 13.4 months to 13% at 50.9 months. Partial responses were observed in 52% of patients; 21% had stable disease, and 12% had progressive disease. Median time to response was 2.8 months (range 1.1-37.1 months), and median duration of response was not reached (range 1.4+ to 58.0+ months). At data cut-off, 37 (48%) patients had ongoing responses. Median progression-free survival was not reached [95% CI 38.4 months-not estimable (NE)], and median overall survival was not reached (95% CI NE). Grade 3-4 treatment-related adverse events (TRAEs) were observed in 32% of patients; 13% of patients had any-grade TRAEs leading to discontinuation. CONCLUSIONS: The results confirm long-term benefit of nivolumab plus low-dose ipilimumab for previously treated patients with MSI-H/dMMR mCRC. The safety profile was manageable with no new safety signals.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Reparación de la Incompatibilidad de ADN/genética , Estudios de Seguimiento , Humanos , Ipilimumab , Inestabilidad de Microsatélites , Nivolumab/uso terapéutico
2.
J Immunother Cancer ; 7(1): 84, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30917871

RESUMEN

BACKGROUND: Prognostic scoring systems are used to estimate the risk of mortality from metastatic renal cell carcinoma (mRCC). Outcomes from different therapies may vary within each risk group. These survival algorithms have been applied to assess outcomes in patients receiving T-cell checkpoint inhibitory immunotherapy and tyrosine kinase inhibitor therapy, but have not been applied extensively to patients receiving high dose interleukin-2 (HD IL-2) immunotherapy. METHODS: Survival of 810 mRCC patients treated from 2006 to 2017 with high dose IL-2 (aldesleukin) and enrolled in the PROCLAIMSM registry data base was assessed utilizing the International Metastatic RCC Database Consortium (IMDC) risk criteria. Median follow-up is 23.4 months (mo.) (range 0.2-124 mo.). Subgroup evaluations were performed by separating patients by prior or no prior therapy, IL-2 alone, or therapy subsequent to IL-2. Some patients were in two groups. We will focus on the 356 patients who received IL-2 alone, and evaluate outcome by risk factor categories. RESULTS: Among the 810 patients, 721 were treatment-naïve (89%) and 59% were intermediate risk. Overall, of the 249 patients with favorable risk, the median overall survival (OS) is 63.3 mo. and the 2-year OS is 77.6%. Of 480 patients with intermediate risk, median OS is 42.4 mo., 2-year OS 68.2%, and of 81 patients with poor risk, median OS 14 mo., 2-year OS 40.4%. Among those who received IL-2 alone (356 patients), median OS is 64.5, 57.6, and 14 months for favorable, intermediate and poor risk categories respectively. Two year survival among those treated only with HD IL-2 is 73.4, 63.7 and 39.8%, for favorable, intermediate and poor risk categories respectively. CONCLUSIONS: Among mRCC patients treated with HD IL-2, all risk groups have median and 2-year survival consistent with recent reports of checkpoint or targeted therapies for mRCC. Favorable and intermediate risk (by IMDC) patients treated with HD IL-2 have longer OS compared with poor risk patients, with most durable OS observed in favorable risk patients. Favorable risk patients treated with HD IL-2 alone have a 2-year OS of 74%. These data continue to support a recommendation for HD IL-2 for patients with mRCC who meet eligibility criteria. TRIAL REGISTRATION: PROCLAIM, NCT01415167 was registered with ClinicalTrials.gov on August 11, 2011, and initiated for retrospective data collection until 2006, and prospective data collection ongoing since 2011.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-2/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Anciano , Antineoplásicos/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Interleucina-2/uso terapéutico , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Estudios Prospectivos , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
3.
Clin Transl Oncol ; 21(6): 721-728, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30374838

RESUMEN

BACKGROUND: Advanced non-small cell lung cancer (NSCLC) has remained challenging to treat effectively. This study aimed to investigate the clinical effects and safety of immunotherapy with dendritic cells and cytokine-induced killer cells (DC-CIK) administered with chemotherapy (CT) in this malignancy. METHODS: We have developed a new clinical trial design termed as the prospective patient's preference-based study (PPPS). Consecutive patients (n = 135) with advanced NSCLC were treated with DC-CIK administered with CT or mono-therapy (CT or DC-CIK alone). RESULTS: For all the patients, the median PFS was 5.7 months and the median OS was 17.5 months. The 1-year PFS and OS rates were 29.4% and 58.2%, respectively. The 1-year PFS and OS rates for DC-CIK plus CT were significantly higher than that in the group of patients who received DC-CIK alone and CT alone (P < 0.05). The number of adoptively infused DC-CIK cells was associated with clinical efficacy. After adjusting for competing risk factors, DC-CIK combined with CT and infused number of CIKs remained independent predictors of PFS and OS. Phenotypic analysis of peripheral blood mononuclear cells showed that CD8+CD28+, and CD8+CD28- T cells, changed significantly in all groups (P < 0.01). The CD3+ T cells increased in the chemotherapy plus immunotherapy and the immunotherapy alone group (P < 0.01), while CD3-CD16+CD56 T cells decreased in the chemotherapy plus immunotherapy and the immunotherapy alone group (P < 0.01). CONCLUSIONS: DC-CIK combined with chemotherapy administration resulted in numerically superior PFS and OS compared with monotherapy in advanced NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Células Asesinas Inducidas por Citocinas/trasplante , Células Dendríticas/trasplante , Inmunoterapia Adoptiva , Neoplasias Pulmonares/terapia , Prioridad del Paciente , Linfocitos T/trasplante , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Células Asesinas Inducidas por Citocinas/inmunología , Células Dendríticas/inmunología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Linfocitos T/inmunología
4.
Br J Dermatol ; 181(3): 580-583, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30244487

RESUMEN

Programmed cell death 1 (PD-1) blockade has rapidly emerged as an effective therapy for a wide variety of metastatic malignancies. It has been associated with multiple immune-related adverse effects, including cutaneous eruptions. We describe two patients with clinical and histological findings that were consistent with subacute cutaneous lupus erythematosus (SCLE) after receiving PD-1 inhibitor therapy for metastatic lung cancer. We successfully treated our first patient with systemic and topical steroids, photoprotection and hydroxychloroquine. However, he subsequently developed dermatomyositis after continuing PD-1 inhibitor therapy. Our second patient presented with a protracted course of a cutaneous eruption in spite of discontinuation of anti-PD-1 therapy and treatment with systemic corticosteroids and infliximab. This patient's SCLE resolved after the addition of topical steroids and photoprotection and discontinuation of anti-tumour necrosis factor therapy. She and her oncology team decided to pursue non-PD-1 inhibitor treatment for lung cancer owing to a lack of tumour response. We add SCLE and dermatomyositis to the growing list of autoimmune complications of PD-1 blockade. Our cases raise a number of questions, particularly in relation to the viability of continuing anti-PD-1 therapy after developing SCLE and the role of immunosuppressive therapy in patients with PD-1 inhibitor-associated connective tissue disease. What's already known about this topic? Programmed cell death 1 (PD-1) blockade, which is rapidly emerging as a therapy for a wide variety of metastatic malignancies, has been associated with multiple immune-related adverse effects. These include systemic autoimmune diseases such as colitis and thyroiditis in addition to numerous cutaneous adverse events. Cutaneous side-effects of PD-1 inhibitors most commonly reported in clinical trials include lichenoid reactions, eczematous dermatitis and vitiligo. What does this study add? We report two cases of PD-1 inhibitor-associated subacute cutaneous lupus erythematosus (SCLE), with one patient progressing to dermatomyositis with continued PD-1 inhibitor treatment. In addition to being a novel cutaneous adverse event, we also demonstrate the possibility of development of multiple autoimmune diseases in one patient, which is different from classic drug-related SCLE. We discuss the treatment challenges for patients with autoimmune skin disease receiving PD-1 inhibitor therapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Dermatomiositis/inmunología , Lupus Eritematoso Cutáneo/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Biopsia , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Dermatomiositis/inducido químicamente , Dermatomiositis/diagnóstico , Dermatomiositis/patología , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Lupus Eritematoso Cutáneo/inducido químicamente , Lupus Eritematoso Cutáneo/diagnóstico , Lupus Eritematoso Cutáneo/patología , Masculino , Persona de Mediana Edad , Nivolumab/efectos adversos , Receptor de Muerte Celular Programada 1/inmunología , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/inmunología
5.
Cell Death Dis ; 7: e2073, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26821068

RESUMEN

Inflammatory breast cancer (IBC) is the deadliest, distinct subtype of breast cancer. High expression of epidermal growth factor receptors [EGFR or human epidermal growth factor receptor 2 (HER2)] in IBC tumors has prompted trials of anti-EGFR/HER2 monoclonal antibodies to inhibit oncogenic signaling; however, de novo and acquired therapeutic resistance is common. Another critical function of these antibodies is to mediate antibody-dependent cellular cytotoxicity (ADCC), which enables immune effector cells to engage tumors and deliver granzymes, activating executioner caspases. We hypothesized that high expression of anti-apoptotic molecules in tumors would render them resistant to ADCC. Herein, we demonstrate that the most potent caspase inhibitor, X-linked inhibitor of apoptosis protein (XIAP), overexpressed in IBC, drives resistance to ADCC mediated by cetuximab (anti-EGFR) and trastuzumab (anti-HER2). Overexpression of XIAP in parental IBC cell lines enhances resistance to ADCC; conversely, targeted downregulation of XIAP in ADCC-resistant IBC cells renders them sensitive. As hypothesized, this ADCC resistance is in part a result of the ability of XIAP to inhibit caspase activity; however, we also unexpectedly found that resistance was dependent on XIAP-mediated, caspase-independent suppression of reactive oxygen species (ROS) accumulation, which otherwise occurs during ADCC. Transcriptome analysis supported these observations by revealing modulation of genes involved in immunosuppression and oxidative stress response in XIAP-overexpressing, ADCC-resistant cells. We conclude that XIAP is a critical modulator of ADCC responsiveness, operating through both caspase-dependent and -independent mechanisms. These results suggest that strategies targeting the effects of XIAP on caspase activation and ROS suppression have the potential to enhance the activity of monoclonal antibody-based immunotherapy.


Asunto(s)
Neoplasias Inflamatorias de la Mama/inmunología , Neoplasias Inflamatorias de la Mama/terapia , Proteína Inhibidora de la Apoptosis Ligada a X/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Línea Celular Tumoral , Cetuximab/farmacología , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunoterapia/métodos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/patología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor ErbB-2/biosíntesis , Trastuzumab/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/deficiencia , Proteína Inhibidora de la Apoptosis Ligada a X/genética
6.
Ann Oncol ; 24(2): 420-428, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23028040

RESUMEN

BACKGROUND: We previously reported results of a prospective trial evaluating the significance of circulating tumor cells (CTCs) in patients with metastatic colorectal cancer (mCRC). This secondary analysis assessed the relationship of the CTC number with carcinoembryonic antigen (CEA) and overall survival. PATIENTS AND METHODS: Patients with mCRC had CTCs measured at baseline and specific time points after the initiation of new therapy. Patients with a baseline CEA value ≥ 10 ng/ml and CEA measurements within ± 30 days of the CTC collection were included. RESULTS: We included 217 patients with mCRC who had a CEA value of ≥ 10 ng/ml. Increased baseline CEA was associated with shorter survival (15.8 versus 20.7 months, P = 0.012). Among all patients with a baseline CEA value of ≥ 25 ng/ml, patients with low baseline CTCs (<3, n = 99) had longer survival than those with high CTCs (≥ 3, n = 58; 20.8 versus 11.7 months, P = 0.001). CTCs added prognostic information at the 3-5- and 6-12-week time points regardless of CEA. In a multivariate analysis, CTCs at baseline but not CEA independently predicted survival and both CTCs and CEA independently predicted survival at 6-12 weeks. CONCLUSIONS: This study demonstrates that both CEA and CTCs contribute prognostic information for patients with mCRC.


Asunto(s)
Antígeno Carcinoembrionario/sangre , Neoplasias Colorrectales , Células Neoplásicas Circulantes , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/mortalidad , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Sobrevida , Adulto Joven
7.
Cancer Gene Ther ; 19(1): 30-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21997230

RESUMEN

CD4+CD25+regulatory T cells (T(reg)) impair anti-tumor and anti-viral immunity. As there are higher T(reg) levels in cancer patients compared with healthy individuals, there is considerable interest in eliminating them or altering their function as part of cancer or viral immunotherapy strategies. The scurfin transcriptional regulator encoded by the member of the forkhead winged helix protein family (FOXP3) is critical for maintaining the functions of T(reg). We hypothesized that targeting FOXP3 expression with a novel arginine-rich, cell-penetrating, peptide-conjugated phosphorodiamidate morpholino (PPMO) based antisense would eliminate T(reg) and enhance the induction of effector T-cell responses. We observed that the PPMO was taken up by activated T cells in vitro and could downregulate FOXP3 expression, which otherwise increases during antigen-specific T-cell activation. Generation of antigen-specific T cells in response to peptide stimulation was enhanced by pre-treatment of peripheral blood mononuclear cells with the FOXP3-targeted PPMO. In summary, modulation of T(reg) levels using the FOXP3 PPMO antisense-based genomic strategy has the potential to optimize immunotherapy strategies in cancer and viral immunotherapy.


Asunto(s)
Factores de Transcripción Forkhead/genética , Morfolinos/farmacología , Oligonucleótidos Antisentido/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Estudios de Casos y Controles , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/inmunología , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Morfolinos/genética , Morfolinos/farmacocinética , Neoplasias/sangre , Neoplasias/inmunología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacocinética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Linfocitos T Reguladores/metabolismo
8.
Clin Exp Immunol ; 163(3): 324-32, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21175594

RESUMEN

Selection of suitable antigens is critical for the development of cancer vaccines. Most desirable are over-expressed cell surface proteins that may serve as targets for both antibodies and T cells, thus maximizing a concerted immune response. Towards this goal, we characterized the relevance of tumour necrosis factor-α-converting enzyme (ADAM17) for such targeted therapeutics. ADAM17 is one of the several metalloproteinases that play a key role in epidermal growth factor receptor (EGFR) signalling and has recently emerged as a new therapeutic target in several tumour types. In the present study, we analysed the expression profile of ADAM17 in a variety of normal and cancer cells of human origin and found that this protein is over-expressed on the surface of several types of cancer cells compared to the normal counterparts. Furthermore, we analysed the presentation of a human leucocyte antigen (HLA)-A2-restricted epitope from ADAM17 protein to specific T cells established from normal donors as well as ovarian cancer patients. Our analysis revealed that the HLA-A2-restricted epitope is processed efficiently and presented by various cancer cells and not by normal cells. Tumour-specific T cell activation results in the secretion of both interferon-γ and granzyme B that can be blocked by HLA-A2 specific antibodies. Collectively, our data present evidence that ADAM17 can be a potential target antigen to devise novel immunotherapeutic strategies against ovarian, breast and prostate cancer.


Asunto(s)
Proteínas ADAM/inmunología , Neoplasias de la Mama/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia , Neoplasias Ováricas/inmunología , Neoplasias de la Próstata/inmunología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Granzimas/metabolismo , Antígeno HLA-A2/inmunología , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/inmunología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Fragmentos de Péptidos/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
9.
Br J Cancer ; 102(1): 124-33, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19953093

RESUMEN

BACKGROUND: Novel technologies to redirect T-cell killing against cancer cells are emerging. We hypothesised that metastatic human colorectal cancer (CRC) previously treated with conventional chemotherapy would be sensitive to T-cell killing mediated by carcinoembryonic antigen (CEA)/CD3-bispecific T-cell-engaging BiTE antibody (MEDI-565). METHODS: We analysed proliferation and lysis of CEA-positive (CEA+) CRC specimens that had survived previous systemic chemotherapy and biologic therapy to determine whether they could be killed by patient T cells engaged by MEDI-565 in vitro. RESULTS: At low concentrations (0.1-1 ng ml(-1)), MEDI-565+ T cells caused reduced proliferation and enhanced apoptosis of CEA+ human CRC specimens. High levels of soluble CEA did not impair killing by redirected T cells and there was no increase in resistance to T-cell killing despite multiple rounds of exposure. CONCLUSIONS: This study shows for the first time that metastatic CRC specimens derived from patients previously treated with conventional chemotherapy can be lysed by patient T cells. Clinical testing of cancer immunotherapies, such as MEDI-565 that result in exposure of tumours to large numbers of T cells, is warranted.


Asunto(s)
Adenocarcinoma/secundario , Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Hepáticas/secundario , Linfocitos T Citotóxicos/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Anticuerpos Biespecíficos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Resistencia a Antineoplásicos , Proteína Ligando Fas/fisiología , Granzimas/fisiología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Compuestos Organoplatinos/farmacología , Oxaliplatino , Receptor fas/fisiología
10.
Ann Oncol ; 20(7): 1223-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19282466

RESUMEN

BACKGROUND: We demonstrated that circulating tumor cell (CTC) number at baseline and follow-up is an independent prognostic factor in metastatic colorectal cancer (mCRC). This analysis was undertaken to explore whether patient and treatment characteristics impact the prognostic value of CTCs. PATIENTS AND METHODS: CTCs were enumerated with immunomagnetic separation from the blood of 430 patients with mCRC at baseline and on therapy. Patients were stratified into unfavorable and favorable prognostic groups based on CTC levels of > or = 3 or <3 CTCs/7.5 ml, respectively. Subgroups were analyzed by line of treatment, liver involvement, receipt of oxaliplatin, irinotecan, or bevacizumab, age, and Eastern Cooperative Oncology Group performance status (ECOG PS). RESULTS: Seventy-one percent of deaths have occurred. Median follow-up for living patients is 25.8 months. For all patients, progression-free survival (PFS) and overall survival (OS) for unfavorable compared with favorable baseline CTCs is shorter (4.4 versus 7.8 m, P = 0.004 for PFS; 9.4 versus 20.6 m, P < 0.0001 for OS). In all patient subgroups, unfavorable baseline CTC was associated with inferior OS (P < 0.001). In patients receiving first- or second-line therapy (P = 0.003), irinotecan (P = 0.0001), having liver involvement (P = 0.002), >/=65 years (P = 0.0007), and ECOG PS of zero (P = 0.04), unfavorable baseline CTC was associated with inferior PFS. CONCLUSION: Baseline CTC count is an important prognostic factor within specific subgroups defined by treatment or patient characteristics.


Asunto(s)
Neoplasias Colorrectales/patología , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Bevacizumab , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Irinotecán , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Valor Predictivo de las Pruebas , Pronóstico , Tasa de Supervivencia , Factores de Tiempo , Resultado del Tratamiento
11.
Cancer Gene Ther ; 16(9): 673-82, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19229288

RESUMEN

Recombinant serotype 5 adenovirus (Ad5) vectors lacking E1 expression induce robust immune responses against encoded transgenes in pre-clinical models, but have muted responses in human trials because of widespread pre-existing anti-adenovirus immunity. Attempts to circumvent Ad5-specific immunity by using alternative serotypes or modifying capsid components have not yielded profound clinical improvement. To address this issue, we explored a novel alternative strategy, specifically reducing the expression of structural Ad5 genes by creating E1 and E2b deleted recombinant Ad5 vectors. Our data show that [E1-, E2b-]vectors retaining the Ad5 serotype are potent immunogens in pre-clinical models despite the presence of significant Ad5-specific immunity, in contrast to [E1-] vectors. These pre-clinical studies with E1 and E2b-deleted recombinant Ad5 vectors suggest that anti-Ad immunity will no longer be a limiting factor, and that clinical trials to evaluate their performance are warranted.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Proteínas E1 de Adenovirus/genética , Proteínas E2 de Adenovirus/genética , Proteínas E3 de Adenovirus/genética , Vacunas contra el Cáncer/inmunología , Vectores Genéticos/inmunología , Proteínas E1 de Adenovirus/inmunología , Proteínas E2 de Adenovirus/inmunología , Proteínas E3 de Adenovirus/inmunología , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/metabolismo , Diferenciación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Eliminación de Gen , Humanos , Células Asesinas Naturales/inmunología , Cinética , Ratones , Ratones Endogámicos C57BL
12.
Dig Liver Dis ; 36(6): 412-8, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15248382

RESUMEN

BACKGROUND: It has been suggested that preoperative biliary drainage increases the risk of infectious complications of pancreaticoduodenectomy. AIMS: The aim of this study was to assess complications related to biliary stents/drains and postoperative morbidity in patients undergoing neoadjuvant chemoradiotherapy for periampullary cancer. PATIENTS: One hundred and eighty-four patients with periampullary neoplasms were prospectively selected for neoadjuvant external beam radiation therapy and 5-fluorouracil-based chemotherapy between 1995 and 2002. METHODS: The data were retrospectively completed and analysed with respect to biliary drainage, efficacy and complications of endoscopic biliary stents and postoperative morbidity. Patients who had undergone a surgical biliary bypass were excluded. RESULTS: Data were completed in 168 patients. One hundred and nineteen patients were treated with endoscopic biliary stents, 18 patients had a percutaneous biliary drain and 31 patients did not require biliary drainage. Hospitalisation for stent-related complications was necessary in 15% of the patients with endoscopic biliary stents. Seventy-two patients underwent pancreaticoduodenectomy. There was no significant difference in the rate of wound infections, intra-abdominal abscesses and overall complications between the groups with and without preoperative biliary drainage. CONCLUSIONS: Postoperative infectious complications are common in patients both with and without preoperative biliary drainage. A statistically significant difference in complication rates was not observed between these groups.


Asunto(s)
Drenaje , Neoplasias Pancreáticas/terapia , Pancreaticoduodenectomía/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Ampolla Hepatopancreática , Antimetabolitos Antineoplásicos/uso terapéutico , Bilis , Quimioterapia Adyuvante , Endoscopía del Sistema Digestivo , Femenino , Fluorouracilo/uso terapéutico , Humanos , Ictericia Obstructiva/etiología , Ictericia Obstructiva/terapia , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Pancreaticoduodenectomía/mortalidad , Cuidados Preoperatorios , Estudios Prospectivos , Radioterapia Adyuvante , Estudios Retrospectivos , Stents
13.
Expert Opin Biol Ther ; 1(2): 153-8, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11727526

RESUMEN

Clinically effective cancer immunotherapy has been sought for more than 100 years and has been recently applied most successfully in strategies that passively deliver immune effectors such as monoclonal antibodies (anti-CD20 for lymphoma and anti-HER2/neu for breast cancer), donor lymphocyte infusions in chronic myelongenous leukemia and non-myeloablative allogeneic peripheral blood progenitor transplants for renal cell carcinoma. There is mounting enthusiasm for strategies employing active stimulation of antitumour immune responses. These include vaccines based on tumour antigen proteins and peptides, autologous, allogeneic or gene-modified tumour cells, dendritic cells and antigen-encoding viral vector constructs. Indeed, randomised Phase III clinical trials of autologous tumour cell vaccines for colorectal cancer demonstrated an improvement in disease free survival and a trend toward improved overall survival [1]. Despite these preliminary successes, it is clear that the many strategies under development cannot all be evaluated for survival benefit in large clinical trials that require many years, patients and resources to complete. This highlights the need to develop intermediate markers to help prioritise which agents to test in prospective randomised Phase III trials.


Asunto(s)
Biomarcadores/análisis , Inmunoterapia , Monitorización Inmunológica/métodos , Neoplasias/inmunología , Neoplasias/terapia , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Resultado del Tratamiento
14.
Biochem J ; 360(Pt 2): 335-44, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11716761

RESUMEN

Cell adhesion is fundamental to establishing and maintaining the discrete tissues in multicellular organisms. Adhesion must be sufficiently strong to preserve tissue architecture, whilst having the capacity to readily dissociate to permit fundamental processes, such as wound repair, to occur. However, very little is known about the signalling mechanisms involved in temporary down-regulation of cell adhesion to facilitate such processes. Cadherins are the principal mediators of cell-cell adhesion in a wide variety of tissues and species and form multi-protein complexes with cytosolic and cytoskeletal proteins to express their full adhesive capacity. In the present study we report that the p85 subunit of phosphoinositide 3-kinase (PI 3-kinase) is associated with the cadherin-based adhesion complex in human epithelial cells. The interaction of p85 with the complex is via beta-catenin. We also show that the interaction of p85 and beta-catenin is direct, involves the N-terminal Src homology domain 2 of p85 and is regulated by tyrosine phosphorylation. These data suggest that PI 3-kinase may play a role in the functional regulation of the cadherin-based adhesion complex.


Asunto(s)
Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fragmentos de Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transactivadores , Animales , Cadherinas/aislamiento & purificación , Dominio Catalítico , Adhesión Celular , Línea Celular , Precipitación Química , Proteínas del Citoesqueleto/genética , Perros , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Glutatión Transferasa/genética , Humanos , Queratinocitos/enzimología , Queratinocitos/metabolismo , Sustancias Macromoleculares , Fragmentos de Péptidos/aislamiento & purificación , Fosfatidilinositol 3-Quinasas/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Técnicas del Sistema de Dos Híbridos , beta Catenina
15.
Crit Rev Immunol ; 21(1-3): 287-97, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11642610

RESUMEN

Successful application of active immunotherapy to the treatment of cancer will require stimulation of potent antigen-specific T-cell responses. It is not known how numerous or how potent these T cells must be in order to abrogate tumors, but the levels of immunity needed to control chronic viral infections may provide estimates for comparison. Evaluation of the efficacy of a vaccine strategy in attaining these levels of immunity will depend on the use of assays that create a picture of T-cell number and function that correlates with clinical outcomes. We discuss the currently available in vivo and in vitro T-cell assays and their relevance for detecting therapeutic levels of T-cell activity. We also propose a strategy for efficiently evaluating the immunologic efficacy of cancer vaccines so that the most promising candidates can be brought more rapidly into definitive clinical trials.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Linfocitos T/inmunología , Citocinas/análisis , Citocinas/genética , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Hipersensibilidad Tardía , Activación de Linfocitos , ARN Mensajero/análisis
17.
Cancer Res ; 61(16): 6112-9, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11507061

RESUMEN

Fruit and vegetable consumption has consistently been associated with decreased risk of a number of aerodigestive tract cancers, including esophageal cancer. We have taken a "food-based" chemopreventive approach to evaluate the inhibitory potential of lyophilized black raspberries (LBRs) against N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumorigenesis in the F344 rat, during initiation and postinitiation phases of carcinogenesis. Anti-initiation studies included a 30-week tumorigenicity bioassay, quantification of DNA adducts, and NMBA metabolism study. Feeding 5 and 10% LBRs, for 2 weeks prior to NMBA treatment (0.25 mg/kg, weekly for 15 weeks) and throughout a 30-week bioassay, significantly reduced tumor multiplicity (39 and 49%, respectively). In a short-term bioassay, 5 and 10% LBRs inhibited formation of the promutagenic adduct O(6)-methylguanine (O(6)-meGua) by 73 and 80%, respectively, after a single dose of NMBA at 0.25 mg/kg. Feeding 5% LBRs also significantly inhibited adduct formation (64%) after NMBA administration at 0.50 mg/kg. The postinitiation inhibitory potential of berries was evaluated in a second bioassay with sacrifices at 15, 25, and 35 weeks. Administration of LBRs began after NMBA treatment (0.25 mg/kg, three times per week for 5 weeks). LBRs inhibited tumor progression as evidenced by significant reductions in the formation of preneoplastic esophageal lesions, decreased tumor incidence and multiplicity, and reduced cellular proliferation. At 25 weeks, both 5 and 10% LBRs significantly reduced tumor incidence (54 and 46%, respectively), tumor multiplicity (62 and 43%, respectively), proliferation rates, and preneoplastic lesion development. Yet, at 35 weeks, only 5% LBRs significantly reduced tumor incidence and multiplicity, proliferation indices and preneoplastic lesion formation. In conclusion, dietary administration of LBRs inhibited events associated with both the initiation and promotion/progression stages of carcinogenesis, which is promising considering the limited number of chemopreventives with this potential.


Asunto(s)
Neoplasias Esofágicas/prevención & control , Frutas , Animales , Carcinógenos/antagonistas & inhibidores , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Quimioprevención/métodos , Aductos de ADN/antagonistas & inhibidores , Aductos de ADN/biosíntesis , Dieta , Dimetilnitrosamina/análogos & derivados , Dimetilnitrosamina/antagonistas & inhibidores , Dimetilnitrosamina/metabolismo , Dimetilnitrosamina/toxicidad , Ácido Elágico/farmacología , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/metabolismo , Liofilización , Guanina/análogos & derivados , Guanina/antagonistas & inhibidores , Guanina/biosíntesis , Masculino , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/prevención & control , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Endogámicas F344
18.
Curr Opin Mol Ther ; 3(4): 407-12, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11525565

RESUMEN

Therion Biologics, the NCI and Aventis Pasteur are investigating CEA-TRICOM, a recombinant, pox virus-based vaccine that incorporates a triple dose of costimulatory molecules as well as the carcinoembryonic antigen (CEA) tumor antigen, for the potential treatment of colorectal cancer. CEA-TRICOM is designed to stimulate and strengthen the body's immune system to kill colorectal cancer cells. CEA-TRICOM is administered in a priming and boosting protocol using two unique pox virus vectors, rV-CEA-TRICOM (recombinant vaccinia vector) and rF-CEA-TRICOM (recombinant fowlpox vector). The TRICOM component of both rV-CEA-TRICOM and rF-CEA-TRICOM comprises three costimulatory molecule transgenes (B7-1, ICAM-1 and LFA-3) [414643], [414645], known to elicit strong cellular immune responses necessary for complete tumor destruction. In preclinical studies conducted by the NCI and Therion, researchers have demonstrated that this combination of three costimulatory molecules dramatically boosts the immune response to eradicate cancer in murine models [399610], [414631]. In February 2001, Therion Biologics and the NCI initiated a phase I trial of CEA-TRICOM [399610]. The phase I trial of CEA-TRICOM is designed to demonstrate proof-of-principle for using multiple costimulatory molecules in conjunction with a tumor antigen to improve the strength of cellular immune responses. It is a multistage, dose-escalation study that will assess the safety and immunologic effects of CEA-TRICOM in up to 42 patients who have advanced metastatic colorectal cancer. Subjects will receive rF-CEA-TRICOM alone, rV-CEA-TRICOM followed by booster vaccinations with rF-CEA-TRICOM or rV-CEA-TRICOM followed by rF-CEA-TRICOM and GM-CSF adjuvant. The primary measure of immune response will be the level of CEA-specific T-cells stimulated by vaccination, with levels of CEA-expressing tumor cells in the blood used as a potential secondary measure of treatment effect [399610].


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/terapia , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Neoplasias Colorrectales/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Vacunación
19.
Anticancer Res ; 21(3B): 1713-22, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11497251

RESUMEN

This study investigated whether local delivery of 10-hydroxycamptothecin provides effective inductive chemotherapy as assessed by significant tumor reduction. Established tumorigenic human oral squamous cell carcinoma cells were used for these experiments. The experimental groups were comprised of: control (blank (no drug) poly(lactide-co-glycolide) (PLGA) microspheres), intraperitoneal 10-hydroxycamptothecin delivery + blank microspheres, local bolus 10-hydroxycamptothecin + blank microspheres, and PLGA controlled-release microspheres. The 10-hydroxycamptothecin dose administered was 12 mg/kg (bolus-intraperitoneal, local) or controlled-release over 10 days. Regardless of delivery route, 10-hydroxycamptothecin significantly reduces tumor volume. However, PLGA microspheres provide significantly higher intratumor-drug concentrations (approximately 10 and 100 fold higher) relative to local bolus and intraperitoneal routes, respectively. Also, only the PLGA microspheres significantly reduced tumor weights. Camptothecin clinical applications are limited by drug inactivation at physiological pH and the need for sustained infusions. However, due to their acidic, camptothecin-stabilizing microclimate, PLGA microspheres could provide a novel delivery system for camptothecin-based induction chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Camptotecina/administración & dosificación , Camptotecina/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Microesferas , Neoplasias de la Boca/tratamiento farmacológico , Poliglactina 910/química , Animales , Cromatografía Líquida de Alta Presión , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Ácido Láctico/química , Pulmón/patología , Ratones , Ratones SCID , Metástasis de la Neoplasia , Trasplante de Neoplasias , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/química , Factores de Tiempo , Células Tumorales Cultivadas
20.
J Natl Cancer Inst ; 93(13): 990-8, 2001 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-11438564

RESUMEN

BACKGROUND: 9-cis-Retinoic acid (9-cis-RA) and N-(4-hydroxyphenyl)retinamide (4-HPR) are effective chemopreventive agents against epithelial tumors in the oral cavity, breast, and prostate. We tested the inhibitory activity of these retinoids against N-nitrosomethylbenzylamine (NMBA)-induced tumorigenesis in the rat esophagus. METHODS: Male Fischer 344 rats were randomly assigned to receive diets either lacking or containing 9-cis-RA or 4-HPR for 1 week before tumor initiation with NMBA and then for the duration of the study. NMBA metabolism, O(6)-methylguanine adduct formation, and cytochrome P450 messenger RNA (mRNA) expression in the esophagi of the rats were studied to investigate the mechanisms by which dietary 4-HPR affects tumorigenesis. All statistical tests were two-sided. RESULTS: Dietary 4-HPR resulted in a dose-dependent and statistically significant enhancement (P<.05) of tumorigenesis in response to NMBA. In two different tumor bioassays, the mean tumor multiplicity for rats fed the highest concentration of dietary 4-HPR (0.8 g/kg diet) was increased by 5.9 tumors (95% confidence interval [CI] = 1.7 to 10.1 tumors) and 6.7 tumors (95% CI = 5.6 to 7.8 tumors) compared with the mean tumor multiplicity for rats that received the control diet lacking 4-HPR. Animals fed diets containing 9-cis-RA displayed no statistically significant increase in tumorigenesis. Compared with animals fed a diet lacking 4-HPR, animals fed 4-HPR had increased NMBA metabolism in esophageal explant cultures and had higher levels of O(6)-methylguanine DNA adducts and CYP2A3 mRNA in their esophagi. CONCLUSIONS: Dietary 4-HPR enhances tumorigenesis in response to NMBA in the rat esophagus by increasing tumor initiation events. Dietary 4-HPR may exert paradoxical effects at some sites, such as the aerodigestive tract, by modulating the bioactivation of carcinogens in target tissues.


Asunto(s)
Antineoplásicos/uso terapéutico , Dimetilnitrosamina/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Fenretinida/uso terapéutico , Alitretinoína , Animales , Antineoplásicos/administración & dosificación , Carcinógenos , Sistema Enzimático del Citocromo P-450/metabolismo , Aductos de ADN , Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/inducido químicamente , Esófago/efectos de los fármacos , Fenretinida/administración & dosificación , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Retinoides/uso terapéutico , Factores de Tiempo , Tretinoina/administración & dosificación , Tretinoina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...