Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Immunother Cancer ; 10(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35618289

RESUMEN

BACKGROUND: Accumulation of regulatory T cells (Treg) has been described to often correlate with poor prognosis in many solid tumors. How Treg presence impinges on limited functionality and clonal composition of tumor-associated CD8 +T cells has important implications for their therapeutic targeting in the tumor microenvironment. In the present study, we investigated how accumulation of Tregs contributes to T cell dysfunction and clonal constriction of tumor-infiltrating CD8 +T cells. METHODS: Resected melanoma and lung adenocarcinoma tissues from tumor-bearing mice or patients were analyzed. The proportions and phenotype as well as clonal diversity of tumor-associated CD8 +T cells were evaluated by flow cytometry and single-cell T-cell receptor (TCR) sequencing, respectively, at early or advanced tumor stages or under Treg depletion conditions. Furthermore, antigen-specific T cells were evaluated on adoptive transfer into tumor-bearing mice in the presence or absence of anti-CTLA-4 antibody or CTLA-4 Ig. Lastly, tumor-bearing mice were treated with anti-KLRG1 antibody and/or bromodomain inhibitor JQ1 with interleukin (IL)-2 immune complexes to determine therapeutic efficacy. RESULTS: We demonstrate that the emergence of exhaustion-like phenotype and impaired effector functionality in tumor-associated CD8 +T cells is positively correlated with Treg accumulation in the tumor bed and this dysfunctional phenotype becomes reversed on Treg reduction in murine melanoma and lung cancer models. Heightened tumor-associated Treg-expressed CTLA-4 is key to emergence and sustenance of this phenotype. Furthermore, TCR sequencing revealed a clonal shrinkage of tumor-infiltrating CD8 +T cells as tumor progressed, which was associated with reduced survival profile concomitant to increasing Treg proportions. Limited IL-2 availability was a key mechanism contributing to this peripheral repertoire reshaping as Treg depletion improved IL-2 levels, rescued CD8 +T cell viability, and improved their clonal diversity. Finally, targeted reduction of tumor but not peripheral Tregs through JQ1 and/or anti-KLRG1 antibody significantly improved antitumor response in melanoma-bearing mice when supplemented with IL-2 immune complexes. CONCLUSION: Collectively, our study reveals a bimodal program enacted by Tregs to support T cell dysfunction in the tumor bed and highlights a promising therapeutic regimen for localized reprogramming of the tumor microenvironment to curb Treg impairment of antitumor CD8 +T cell response in favor of improved antitumor immunity.


Asunto(s)
Adenocarcinoma , Melanoma , Animales , Complejo Antígeno-Anticuerpo , Linfocitos T CD8-positivos , Antígeno CTLA-4 , Humanos , Interleucina-2 , Ratones , Microambiente Tumoral
2.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34793338

RESUMEN

The clinical utility of histone/protein deacetylase (HDAC) inhibitors in combinatorial regimens with proteasome inhibitors for patients with relapsed and refractory multiple myeloma (MM) is often limited by excessive toxicity due to HDAC inhibitor promiscuity with multiple HDACs. Therefore, more selective inhibition minimizing off-target toxicity may increase the clinical effectiveness of HDAC inhibitors. We demonstrated that plasma cell development and survival are dependent upon HDAC11, suggesting this enzyme is a promising therapeutic target in MM. Mice lacking HDAC11 exhibited markedly decreased plasma cell numbers. Accordingly, in vitro plasma cell differentiation was arrested in B cells lacking functional HDAC11. Mechanistically, we showed that HDAC11 is involved in the deacetylation of IRF4 at lysine103. Further, targeting HDAC11 led to IRF4 hyperacetylation, resulting in impaired IRF4 nuclear localization and target promoter binding. Importantly, transient HDAC11 knockdown or treatment with elevenostat, an HDAC11-selective inhibitor, induced cell death in MM cell lines. Elevenostat produced similar anti-MM activity in vivo, improving survival among mice inoculated with 5TGM1 MM cells. Elevenostat demonstrated nanomolar ex vivo activity in 34 MM patient specimens and synergistic activity when combined with bortezomib. Collectively, our data indicated that HDAC11 regulates an essential pathway in plasma cell biology establishing its potential as an emerging theraputic vulnerability in MM.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Células Plasmáticas/metabolismo , Animales , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Mieloma Múltiple/fisiopatología
3.
Nat Med ; 27(8): 1410-1418, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34385708

RESUMEN

Adoptive cell therapy using tumor-infiltrating lymphocytes (TILs) has shown activity in melanoma, but has not been previously evaluated in metastatic non-small cell lung cancer. We conducted a single-arm open-label phase 1 trial ( NCT03215810 ) of TILs administered with nivolumab in 20 patients with advanced non-small cell lung cancer following initial progression on nivolumab monotherapy. The primary end point was safety and secondary end points included objective response rate, duration of response and T cell persistence. Autologous TILs were expanded ex vivo from minced tumors cultured with interleukin-2. Patients received cyclophosphamide and fludarabine lymphodepletion, TIL infusion and interleukin-2, followed by maintenance nivolumab. The end point of safety was met according to the prespecified criteria of ≤17% rate of severe toxicity (95% confidence interval, 3-29%). Of 13 evaluable patients, 3 had confirmed responses and 11 had reduction in tumor burden, with a median best change of 35%. Two patients achieved complete responses that were ongoing 1.5 years later. In exploratory analyses, we found T cells recognizing multiple types of cancer mutations were detected after TIL treatment and were enriched in responding patients. Neoantigen-reactive T cell clonotypes increased and persisted in peripheral blood after treatment. Cell therapy with autologous TILs is generally safe and clinically active and may constitute a new treatment strategy in metastatic lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Resistencia a Antineoplásicos , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Adulto , Anciano , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia
4.
Cancer Immunol Res ; 9(5): 554-567, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33653716

RESUMEN

Immunotherapy (IT) and targeted therapy (TT) are both effective against melanoma, but their combination is frequently toxic. Here, we investigated whether the sequence of IT (anti-PD-1)→ TT (ceritinib-trametinib or dabrafenib-trametinib) was associated with improved antitumor responses in mouse models of BRAF- and NRAS-mutant melanoma. Mice with NRAS-mutant (SW1) or BRAF-mutant (SM1) mouse melanomas were treated with either IT, TT, or the sequence of IT→TT. Tumor volumes were measured, and samples from the NRAS-mutant melanomas were collected for immune-cell analysis, single-cell RNA sequencing (scRNA-seq), and reverse phase protein analysis (RPPA). scRNA-seq demonstrated that the IT→TT sequence modulated the immune environment, leading to increased infiltration of T cells, monocytes, dendritic cells and natural killer cells, and decreased numbers of tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells. Durable responses to the IT→TT sequence were dependent on T-cell activity, with depletion of CD8+, but not CD4+, T cells abrogating the therapeutic response. An analysis of transcriptional heterogeneity in the melanoma compartment showed the sequence of IT→TT enriched for a population of melanoma cells with increased expression of MHC class I and melanoma antigens. RPPA analysis demonstrated that the sustained immune response induced by IT→TT suppressed tumor-intrinsic signaling pathways required for therapeutic escape. These studies establish that upfront IT improves the responses to TT in BRAF- and NRAS-mutant melanoma models.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Imidazoles/química , Inmunoterapia , Melanoma/genética , Melanoma/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Oximas/química , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Piridonas/química , Pirimidinas/química , Pirimidinonas/química , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Sulfonas/química , Linfocitos T Reguladores/inmunología
5.
Cancer Immunol Immunother ; 67(12): 1853-1862, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30209589

RESUMEN

The GM.CD40L vaccine, which recruits and activates dendritic cells, migrates to lymph nodes, activating T cells and leading to systemic tumor cell killing. When combined with the CCL21 chemokine, which recruits T cells and enhances T-cell responses, additive effects have been demonstrated in non-small cell lung cancer mouse models. Here, we compared GM.CD40L versus GM.CD40L plus CCL21 (GM.CD40L.CCL21) in lung adenocarcinoma patients with ≥ 1 line of treatment. In this phase I/II randomized trial (NCT01433172), patients received intradermal vaccines every 14 days (3 doses) and then monthly (3 doses). A two-stage minimax design was used. During phase I, no dose-limiting toxicities were shown in three patients who received GM.CD40L.CCL21. During phase II, of evaluable patients, 5/33 patients (15.2%) randomized for GM.DCD40L (p = .023) and 3/32 patients (9.4%) randomized for GM.DCD40L.CCL21 (p = .20) showed 6-month progression-free survival. Median overall survival was 9.3 versus 9.5 months with GM.DCD40L versus GM.DCD40L.CCL21 (95% CI 0.70-2.25; p = .44). For GM.CD40L versus GM.CD40L.CCL21, the most common treatment-related adverse events (TRAEs) were grade 1/2 injection site reaction (51.4% versus 61.1%) and grade 1/2 fatigue (35.1% versus 47.2%). Grade 1 immune-mediated TRAEs were isolated to skin. No patients showed evidence of pseudo-progression or immune-related TRAEs of grade 1 or greater of pneumonitis, endocrinopathy, or colitis, and none discontinued treatment due to toxicity. Although we found no significant associations between vaccine immunogenicity and outcomes, in limited biopsies, one patient treated with GMCD40L.CCL21 displayed abundant tumor-infiltrating lymphocytes. This possible effectiveness warrants further investigation of GM.CD40L in combination approaches.


Asunto(s)
Adenocarcinoma/terapia , Ligando de CD40/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/terapia , Quimiocina CCL21/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Inmunoterapia , Adenocarcinoma/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
7.
Neoplasia ; 19(7): 530-536, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28582704

RESUMEN

BACKGROUND: Therapeutic strategies targeting immune checkpoint proteins have led to significant responses in patients with various tumor types. The success of these studies has led to the development of various antibodies/inhibitors for the different checkpoint proteins involved in immune evasion of the tumor. Adenosine present in high concentrations in the tumor microenvironment activates the immune checkpoint adenosine A2a receptor (A2aR), leading to the suppression of antitumor responses. Inhibition of this checkpoint has the potential to enhance antitumor T-cell responsiveness. METHODS: We developed a novel A2aR antagonist (PBF-509) and tested its antitumor response in vitro, in a mouse model, and in non-small cell lung cancer patient samples. RESULTS: Our studies showed that PBF-509 is highly specific to the A2aR as well as inhibitory of A2aR function in an in vitro model. In a mouse model, we found that lung metastasis was decreased after treatment with PBF-509 compared with its control. Furthermore, freshly resected tumor-infiltrating lymphocytes from lung cancer patients showed increased A2aR expression in CD4+ cells and variable expression in CD8+ cells. Ex vivo studies showed an increased responsiveness of human tumor-infiltrating lymphocytes when PBF-509 was combined with anti-PD-1 or anti-PD-L1. CONCLUSIONS: Our studies demonstrate that inhibition of the A2aR using the novel inhibitor PBF-509 could lead to novel immunotherapeutic strategies in non-small cell lung cancer.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor de Adenosina A2A/metabolismo , Microambiente Tumoral/inmunología , 5'-Nucleotidasa/metabolismo , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Metástasis de la Neoplasia , Neoplasias/patología
8.
New Dir Child Adolesc Dev ; 2017(155): 117-130, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28267292

RESUMEN

The Government of Mozambique has long struggled to improve the low reading levels of children in early grades. With funding from the U.S. Agency for International Development (USAID) in 2012, World Education collaborated with the Ministry of Education and Human Development (MINEDH) to improve reading by developing a research-based reading intervention and testing it in two provinces. This article examines student reading performance from cohorts of second and third graders before and after a 1-year intervention compared to that of a control group and identifies factors required for successful scale-up.


Asunto(s)
Evaluación Educacional , Desarrollo de Programa , Lectura , Instituciones Académicas , Niño , Humanos , Mozambique
9.
Clin Cancer Res ; 22(16): 4119-32, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26964571

RESUMEN

PURPOSE: A significant limitation of checkpoint blockade immunotherapy is the relatively low response rate (e.g., ∼20% with PD-1 blockade in lung cancer). In this study, we tested whether strategies that increase T-cell infiltration to tumors can be efficacious in enhancing immunotherapy response. EXPERIMENTAL DESIGN: We performed an unbiased screen to identify FDA-approved oncology agents with an ability to enhance T-cell chemokine expression with the goal of identifying agents capable of augmenting immunotherapy response. Identified agents were tested in multiple lung tumor models as single agents and in combination with PD-1 blockade. Additional molecular and cellular analysis of tumors was used to define underlying mechanisms. RESULTS: We found that histone deacetylase (HDAC) inhibitors (HDACi) increased expression of multiple T-cell chemokines in cancer cells, macrophages, and T cells. Using the HDACi romidepsin in vivo, we observed increased chemokine expression, enhanced T-cell infiltration, and T-cell-dependent tumor regression. Importantly, romidepsin significantly enhanced the response to PD-1 blockade immunotherapy in multiple lung tumor models, including nearly complete rejection in two models. Combined romidepsin and PD-1 blockade also significantly enhanced activation of tumor-infiltrating T cells. CONCLUSIONS: These results provide evidence for a novel role of HDACs in modulating T-cell chemokine expression in multiple cell types. In addition, our findings indicate that pharmacologic induction of T-cell chemokine expression represents a conceptually novel approach for enhancing immunotherapy response. Finally, these results suggest that combination of HDAC inhibitors with PD-1 blockade represents a promising strategy for lung cancer treatment. Clin Cancer Res; 22(16); 4119-32. ©2016 AACR.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/inmunología , Quimiocinas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/fisiología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Modelos Biológicos , Terapia Molecular Dirigida , Mutación , Resultado del Tratamiento , Carga Tumoral
10.
Oncotarget ; 7(16): 22928-38, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27008709

RESUMEN

PURPOSE: Indoximod is an oral inhibitor of the indoleamine 2,3-dioxygenase pathway, which causes tumor-mediated immunosuppression. Primary endpoints were maximum tolerated dose (MTD) and toxicity for indoximod in patients with advanced solid tumors. Secondary endpoints included response rates, pharmacokinetics, and immune correlates. EXPERIMENTAL DESIGN: Our 3+3 phase I trial comprised 10 dose levels (200, 300, 400, 600, and 800 mg once/day; 600, 800, 1200, 1600, and 2000 mg twice/day). Inclusion criteria were measurable metastatic solid malignancy, age ≥18 years, and adequate organ/marrow function. Exclusion criteria were chemotherapy ≤ 3 weeks prior, untreated brain metastases, autoimmune disease, or malabsorption. RESULTS: In 48 patients, MTD was not reached at 2000 mg twice/day. At 200 mg once/day, 3 patients previously treated with checkpoint inhibitors developed hypophysitis. Five patients showed stable disease >6 months. Indoximod plasma AUC and Cmax plateaued above 1200mg. Cmax (~12 µM at 2000 mg twice/day) occurred at 2.9 hours, and half-life was 10.5 hours. C reactive protein (CRP) levels increased across multiple dose levels. CONCLUSIONS: Indoximod was safe at doses up to 2000 mg orally twice/day. Best response was stable disease >6 months in 5 patients. Induction of hypophysitis, increased tumor antigen autoantibodies and CRP levels were observed.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Neoplasias/tratamiento farmacológico , Triptófano/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Triptófano/administración & dosificación , Triptófano/efectos adversos , Adulto Joven
11.
BMC Cancer ; 16: 176, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26935219

RESUMEN

BACKGROUND: Anti-fibrotic drugs such as pirfenidone have been developed for the treatment of idiopathic pulmonary fibrosis. Because activated fibroblasts in inflammatory conditions have similar characteristics as cancer-associated fibroblasts (CAFs) and CAFs contribute actively to the malignant phenotype, we believe that anti-fibrotic drugs have the potential to be repurposed as anti-cancer drugs. METHODS: The effects of pirfenidone alone and in combination with cisplatin on human patient-derived CAF cell lines and non-small cell lung cancer (NSCLC) cell lines were examined. The impact on cell death in vitro as well as tumor growth in a mouse model was determined. Annexin V/PI staining and Western blot analysis were used to characterize cell death. Synergy was assessed with the combination index method using Calcusyn software. RESULTS: Pirfenidone alone induced apoptotic cell death in lung CAFs at a high concentration (1.5 mg/mL). However, co-culture in vitro experiments and co-implantation in vivo experiments showed that the combination of low doses of cisplatin (10 µM) and low doses of pirfenidone (0.5 mg/mL), in both CAFs and tumors, lead to increased cell death and decreased tumor progression, respectively. Furthermore, the combination of cisplatin and pirfenidone in NSCLC cells (A549 and H157 cells) leads to increased apoptosis and synergistic cell death. CONCLUSIONS: Our studies reveal for the first time that the combination of cisplatin and pirfenidone is active in preclinical models of NSCLC and therefore may be a new therapeutic approach in this disease.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Neoplasias/patología , Piridonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Fibroblastos/patología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Br J Hosp Med (Lond) ; 74(10): 577-80, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24105311

RESUMEN

This article gives a practical guide for the management of open lower limb fractures. It outlines the referral criteria and pathway for definitive care in a specialist centre, the initial management steps that should be taken in the emergency department, and the principles of fixation, soft tissue coverage and antibiotic therapy.


Asunto(s)
Fracturas Abiertas/terapia , Traumatismos de la Pierna/terapia , Infección de Heridas/prevención & control , Amputación Quirúrgica , Antibacterianos/uso terapéutico , Desbridamiento , Fijación Interna de Fracturas , Fracturas Abiertas/etiología , Fracturas Abiertas/patología , Humanos , Traumatismos de la Pierna/etiología , Traumatismos de la Pierna/patología , Traumatismos de los Tejidos Blandos/etiología , Traumatismos de los Tejidos Blandos/patología , Traumatismos de los Tejidos Blandos/terapia , Colgajos Quirúrgicos , Infección de Heridas/etiología , Infección de Heridas/patología
13.
J Immunother ; 36(8): 442-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23994887

RESUMEN

We created a vaccine in which irradiated allogeneic lung adenocarcinoma cells are combined with a bystander K562 cell line transfected with hCD40L and hGM-CSF. By recruiting and activating dendritic cells, we hypothesized that the vaccine would induce tumor regression in metastatic lung adenocarcinoma. Intradermal vaccine was given q14 days×3, followed by monthly ×3. Cyclophosphamide (300 mg/m IV) was administered before the first and fourth vaccines to deplete regulatory T cells. All-trans retinoic acid was given (150/mg/m/d) after the first and fourth vaccines to enhance dendritic cell differentiation. Twenty-four participants were accrued at a single institution from October 2006 to June 2008, with a median age 64 years and median of 4 previous lines of systemic therapy. A total of 101 vaccines were administered. Common toxicities were headache (54%) and site reaction (38%). No radiologic responses were observed. Median overall survival was 7.9 months and median progression-free survival was 1.7 months. Of 14 patients evaluable for immunological study, 5 had peptide-induced CD8 T-cell activation after vaccination. Overall, vaccine administration was feasible in an extensively pretreated population of metastatic lung cancer. Despite a suggestion of clinical activity in the subset with immune response, the trial did not meet the primary endpoint of inducing radiologic tumor regression.


Asunto(s)
Adenocarcinoma/terapia , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Neoplasias Pulmonares/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Anciano , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Efecto Espectador , Ligando de CD40/genética , Ligando de CD40/metabolismo , Diferenciación Celular/efectos de los fármacos , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Células K562 , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia , Análisis de Supervivencia , Transgenes/genética , Resultado del Tratamiento , Tretinoina/administración & dosificación
14.
Cancer Biol Ther ; 14(9): 860-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23917542

RESUMEN

Recently it has become clear that the cost associated with the Warburg effect, which is inefficient production of ATP, is offset by selective advantages that are produced by resultant intracellular metabolic alterations. In fact tumors may be addicted to the Warburg effect. In addition these alterations result in changes in the extracellular tumor microenvironment that can also produce selective advantages for tumor cell growth and survival. One such extracellular alteration is increased adenosine concentrations that have been shown to impair T cell mediated rejection and support angiogenesis. The expression of the A2A receptor in non-small cell cancer (NSCLC) tissues, cell lines and cancer associated fibroblasts (CAF) was determined by performing immunohistrochemistry and immunoblot analysis. The efficacy of the A2A receptor antagonists in vivo was evaluated in a PC9 xenograft model. To determine the mode of cell death induced by A2A receptor antagonists flow cytometry, immunoblot, and cytotoxic analysis were performed. We found that a significant number of lung adenocarcinomas express adenosine A2A receptors. Antagonism of these receptors impaired CAF and tumor cell growth in vitro and inhibited human tumor xenograft growth in mice. These observations add to the rationale for testing adenosine A2A receptor antagonists as anticancer therapeutics. Not only could there be prevention of negative signaling in T cells within the tumor microenvironment and inhibition of angiogenesis, but also an inhibitory effect on tumor-promoting, immunosuppressive CAFs and a direct inhibitory effect on the tumor cells themselves.


Asunto(s)
Adenocarcinoma/patología , Antagonistas del Receptor de Adenosina A2/farmacología , Proliferación Celular/efectos de los fármacos , Fibroblastos/metabolismo , Neoplasias Pulmonares/patología , Receptor de Adenosina A2A/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Fibroblastos/efectos de los fármacos , Xenoinjertos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones Desnudos , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Receptor de Adenosina A2A/genética , Triazinas/farmacología , Triazinas/uso terapéutico , Triazoles/farmacología , Triazoles/uso terapéutico
15.
Int J Radiat Oncol Biol Phys ; 82(2): 924-32, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21398051

RESUMEN

PURPOSE: The goal of this study was to determine the effect of combination of intratumoral administration of dendritic cells (DC) and fractionated external beam radiation (EBRT) on tumor-specific immune responses in patients with soft-tissue sarcoma (STS). METHODS AND MATERIAL: Seventeen patients with large (>5 cm) high-grade STS were enrolled in the study. They were treated in the neoadjuvant setting with 5,040 cGy of EBRT, split into 28 fractions and delivered 5 days per week, combined with intratumoral injection of 10(7) DCs followed by complete resection. DCs were injected on the second, third, and fourth Friday of the treatment cycle. Clinical evaluation and immunological assessments were performed. RESULTS: The treatment was well tolerated. No patient had tumor-specific immune responses before combined EBRT/DC therapy; 9 patients (52.9%) developed tumor-specific immune responses, which lasted from 11 to 42 weeks. Twelve of 17 patients (70.6%) were progression free after 1 year. Treatment caused a dramatic accumulation of T cells in the tumor. The presence of CD4(+) T cells in the tumor positively correlated with tumor-specific immune responses that developed following combined therapy. Accumulation of myeloid-derived suppressor cells but not regulatory T cells negatively correlated with the development of tumor-specific immune responses. Experiments with (111)In labeled DCs demonstrated that these antigen presenting cells need at least 48 h to start migrating from tumor site. CONCLUSIONS: Combination of intratumoral DC administration with EBRT was safe and resulted in induction of antitumor immune responses. This suggests that this therapy is promising and needs further testing in clinical trials design to assess clinical efficacy.


Asunto(s)
Células Dendríticas/trasplante , Sarcoma/terapia , Neoplasias de los Tejidos Blandos/terapia , Adulto , Anciano , Anciano de 80 o más Años , Movimiento Celular , Terapia Combinada/métodos , Células Dendríticas/diagnóstico por imagen , Células Dendríticas/fisiología , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Inmunidad Humoral/inmunología , Radioisótopos de Indio , Proteínas Inhibidoras de la Apoptosis/inmunología , Inyecciones Intralesiones , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Células Mieloides/inmunología , Terapia Neoadyuvante/métodos , Proteínas de Neoplasias/inmunología , Cintigrafía , Sarcoma/inmunología , Sarcoma/patología , Sarcoma/radioterapia , Neoplasias de los Tejidos Blandos/inmunología , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/radioterapia , Survivin , Linfocitos T/inmunología , Factores de Tiempo
16.
J Immunother ; 31(1): 72-80, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18157014

RESUMEN

We report a single center phase II trial of sequential vaccination followed with vaccine plus interleukin-2 (IL-2). Vaccination consisted of autologous cells cultured from primary tumor or resected metastasis, transduced to express B7.1 surface molecule and then irradiated. The vaccine would hypothetically costimulate tumor-reactive T cells before IL-2 exposure. Treatment plan was 3 subcutaneous vaccine injections at 4-week intervals and subcutaneous IL-2 treatment for 6 weeks starting at week 7. Sixty-six patients enrolled, of whom 39 received at least 1 vaccine; most observed toxicity was attributable to IL-2 not vaccine; best responses were 3% pathologic complete response, 5% partial response, 64% stable disease, and 28% disease progression. Median survival was 21.8 months (95% confidence interval 17.8 to 29.6). Significant postvaccination increases in IFN-gamma responses to autologous tumor were observed in 2/26 cases. Eighty-one percent of posttreatment subdermal delayed-type hypersensitivity tests (using nontransduced, irradiated autologous tumor cells) had biopsies demonstrating injection site lymphocytic infiltration. Post hoc comparison of the median survival of subjects whose biopsies had lymphocytic infiltration appears longer than in the 19% noninfiltrated (28.4 vs. 17.8 mo, P=0.045, two-sided log-rank test). The single arm design precludes conclusive comparison of objective response rates (not different here) or median survival (longer here) versus those of historical series using similar IL-2 schedules alone. Better outcomes could be logically associated to vaccine response (detectable lymphocytic infiltrates) or to random events that a single arm study design cannot address. This vaccine approach may merit further clinical development.


Asunto(s)
Antígeno B7-1/inmunología , Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/terapia , Interleucina-2/uso terapéutico , Neoplasias Renales/terapia , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-1/genética , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-2/efectos adversos , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Análisis de Supervivencia , Transfección , Resultado del Tratamiento , Células Tumorales Cultivadas
17.
Ann Surg Oncol ; 14(2): 869-84, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17103257

RESUMEN

BACKGROUND: Significant antitumor T-cell responses are generated in vitro when human lymphocytes are stimulated with autologous tumor cells in the presence of bystander cells transfected with CD40L and GM-CSF. Our goal was to test this bystander-based vaccine strategy in vivo in cancer patients with stage IV disease. METHODS: Patients received three intradermal vaccine injections (irradiated autologous tumor cells plus GM.CD40L bystander cells) at 28-day intervals. Patients with no disease progression received three additional vaccines at 4, 12, and 24 months. Patients were monitored for toxicity, tumor response, and tumor-specific immune responses. RESULTS: Twenty-one patients received at least three vaccine injections, with no toxicity attributable to the vaccine. Immunohistochemistry of vaccine injection site biopsies with CD1a and CD86 antibodies confirmed recruitment and activation of dendritic cells. There was no tumor regression after vaccination, but many patients had stable disease, including six of ten melanoma patients. Four patients developed tumor-specific T-cell responses on ELISPOT testing. One patient, who had stable disease for 24 months, demonstrated an increase in MART-1-specific T-cells by tetramer analysis after re-immunization; biopsy of the tumor that progressed 2 years after the onset of vaccination revealed a massive peritumoral and intratumoral T-cell infiltrate. CONCLUSIONS: Vaccination of cancer patients with autologous tumor cells and GM.CD40L bystander cells (engineered to express GM-CSF and CD40L) is safe, can recruit and activate dendritic cells, and can elicit tumor-specific T-cell responses. Phase-II trials are underway to evaluate the impact of bystander-based vaccines on melanoma and mantle cell lymphoma.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Ligando de CD40/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias/terapia , Adyuvantes Inmunológicos/fisiología , Adulto , Anciano , Efecto Espectador/inmunología , Ligando de CD40/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inmunoterapia , Leucemia Eritroblástica Aguda , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T/inmunología
18.
Spine (Phila Pa 1976) ; 31(18): E636-40, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16915079

RESUMEN

STUDY DESIGN: A prospective case controlled study to compare the clinical and radiographic performance of Healos soaked in bone marrow aspirate (BMA) to iliac crest autograft when used in lumbar spinal fusion. OBJECTIVE: To evaluate the null hypothesis: Healos used with BMA is not an effective alternative to iliac crest autograft in lumbar spine fusions. SUMMARY OF BACKGROUND DATA: Healos (a Type 1 collagen/hydroxyapatite matrix) is osteoconductive and when soaked for at least 20 minutes in BMA becomes osteoinductive. It is nontoxic and straightforward to use, avoiding the morbidity of autograft harvest. Animal studies and early clinical series in humans have suggested that Healos and BMA are an effective substitute for autograft in certain circumstances. METHODS: From July 2000, Healos and BMA were used as the graft material, instead of autograft harvested from the iliac crest, in all patients undergoing lumbar spinal fusion. Clinical outcome measures used were the Low Back Outcome Score (LBOS), a Patient Satisfaction Score, and the Prolo Economic Score (after Schnee). Standing anteroposterior and lateral radiographs were taken at 12- and 24-month follow-up visits. The first 50 cases in this consecutive series were age, sex, and operative intervention matched to historical controls who underwent surgery between 1997 and 2000 and in whom autograft from the iliac crest had been used as the graft material. Surgical outcome data in these patients had also been gathered prospectively. An independent radiologist, blinded to the graft material, using standard plain radiograph criteria for fusion, examined all the radiographs. An independent surgeon assessed clinical outcomes. RESULTS: For posterolateral lumbar fusions, there were equivalent radiologic fusion rates for the 2 groups with no significant difference in the subjective and objective clinical outcomes. The radiologic fusions rate was significantly lower when Healos had been used for lumbar interbody fusions. Clinical outcomes for both groups were similar. There were no lasting complications associated with Healos use compared with a 14%; persisting donor site complication rate in the autograft patients. CONCLUSION: The null hypothesis is only partially correct. Healos and BMA are not inferior to autologous iliac crest bone as a graft material in posterolateral lumbar spine fusions but are radiographically ineffective in lumbar interbody fusions.


Asunto(s)
Sustitutos de Huesos/administración & dosificación , Implantes Experimentales , Vértebras Lumbares/cirugía , Fusión Vertebral/instrumentación , Trasplante de Médula Ósea , Estudios de Casos y Controles , Femenino , Humanos , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Masculino , Persona de Mediana Edad , Fusión Vertebral/métodos , Resultado del Tratamiento
19.
Fish Shellfish Immunol ; 20(1): 40-6, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15927484

RESUMEN

Reactive nitrogen intermediates, such as nitric oxide (NO), are important immunomodulators in vertebrate immune systems, but have yet to be identified as mediators of host defence in any member of class Chondrichthyes, the cartilaginous fishes. In the present study, production of NO by nurse shark (Ginglymostoma cirratum) peripheral blood leucocytes (PBL) stimulated with bacterial cell wall lipopolysaccharide (LPS) was investigated. PBL were cultured for 24 to 96 h following stimulation with LPS at concentrations ranging from 0 to 25 microg ml(-1), in both serum-supplemented and serum-free culture conditions. Production of NO was measured indirectly using the Griess reaction, with maximal NO production occurring after 72 h using 10% FBS and 10 microg LPS ml(-1). Application of these culture conditions to PBL from another cartilaginous fish (clearnose skate, Raja eglanteria) resulted in a similar NO response. Addition of a specific inhibitor of inducible nitric oxide synthase (iNOS), L-N(6)-(1-iminoethyl)lysine (L-NIL), resulted in a significant decrease in the production of NO by PBL from both species.


Asunto(s)
Leucocitos/metabolismo , Óxido Nítrico/metabolismo , Tiburones/metabolismo , Rajidae/metabolismo , Análisis de Varianza , Animales , Colorimetría , Florida , Lipopolisacáridos , Lisina/análogos & derivados , Lisina/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Nitritos/metabolismo
20.
Integr Comp Biol ; 46(6): 1072-1081, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19343108

RESUMEN

SYNOPSIS: Reports that elasmobranchs (sharks, skates, and rays) may have a low incidence of disease have stimulated interest in understanding the role of their immune system in this apparent resistance. Although research in this area may potentially translate into applications for human health, a basic understanding of the elasmobranch immune system components and how they function is essential. As in higher vertebrates, elasmobranch fishes possess thymus and spleen, but in the absence of bone marrow and lymph nodes, these fish have evolved unique lymphomyeloid tissues, namely epigonal and Leydig organs. As conditions for short-term culture of elasmobranch immune cells have become better understood, the opportunity to examine functional activity of cytokine-like factors derived from conditioned culture medium has resulted in the identification of growth inhibitory activity against a variety of tumor cell lines. Specifically, the medium enriched by short term culture of bonnethead shark (Sphyrna tiburo) epigonal cells (epigonal conditioned medium, ECM) has been shown to inhibit the growth of mammalian tumor cell lines, including fibrosarcoma (WEHI-164), melanoma (A375.S2), B-cell lymphoma (Daudi), T-cell leukemia (Jurkat), pancreatic cancer (PANC-1), ovarian cancer (NIH:OVCAR-3), and three breast carcinoma cell lines (MCF7, HCC38, Hs578T). Of the cell lines tested, WEHI-164, A375.S2, Daudi, and Jurkat cells were among the most sensitive to growth inhibitory activity of ECM whereas PANC-1 and NIH:OVCAR-3 cells were among the least sensitive. In addition, ECM demonstrated preferential growth inhibition of malignant cells in assays against two different malignant/non-malignant cell line pairs (HCC38/HCC38 BL and Hs 578T/Hs 578Bst). Separation of protein components of ECM using SDS-PAGE resulted in a very reproducible pattern of three major bands corresponding to molecular sizes of approximately 40-42 kD, 24 kD, and 17 kD. Activity is lost after heating at 75 degrees C for 30 min, and can be diminished by treatment with proteinase K and protease. Activity is not affected by treating with trypsin, DNase I or RNase A.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...