Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Transl Med ; 17(1): 219, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31288845

RESUMEN

BACKGROUND: The pleiotropic cytokine, transforming growth factor (TGF)-ß, and CD4+CD25+Foxp3+ regulatory T cells (Tregs) play a critical role in actively suppressing antitumor immune responses. Evidence shows that TGF-ß produced by tumor cells promotes tolerance via expansion of Tregs. Our group previously demonstrated that blockade of TGF-ß signaling with a small molecule TGF-ß receptor I antagonist (SM16) inhibited primary and metastatic tumor growth in a T cell dependent fashion. In the current study, we evaluated the effect of SM16 on Treg generation and function. METHODS: Using BALB/c, FoxP3eGFP and Rag-/- mice, we performed FACS analysis to determine if SM16 blocked de novo TGF-ß-induced Treg generation in vitro and in vivo. CD4+ T cells from lymph node and spleen were isolated from control mice or mice maintained on SM16 diet, and flow cytometry analysis was used to detect the frequency of CD4+CD25-FoxP3+ and CD4+CD25+FoxP3+ T cells. In vitro suppression assays were used to determine the ability to suppress naive T cell proliferation in vitro of both CD4+CD25+FoxP3+ and CD4+CD25-FoxP3+ T cell sub-populations. We then examined whether SM16 diet exerted an inhibitory effect on primary tumor growth and correlated with changes in FoxP3+expression. ELISA analysis was used to measure IFN-γ levels after 72 h co-culture of CD4+CD25+ T cells from tumor-bearing mice on control or SM16 diet with CD4+CD25- T cells from naive donors. RESULTS: SM16 abrogates TGF-ß-induced Treg generation in vitro but does not prevent global homeostatic expansion of CD4+ T cell sub-populations in vivo. Instead, SM16 treatment causes expansion of a population of CD4+CD25-Foxp3+ Treg-like cells without significantly altering the overall frequency of Treg in lymphoreplete naive and tumor-bearing mice. Importantly, both the CD4+CD25-Foxp3+ T cells and the CD4+CD25+Foxp3+ Tregs in mice receiving SM16 diet exhibited diminished ability to suppress naive T cell proliferation in vitro compared to Treg from mice on control diet. CONCLUSIONS: These findings suggest that blockade of TGF-ß signaling is a potentially useful strategy for blunting Treg function to enhance the anti-tumor response. Our data further suggest that the overall dampening of Treg function may involve the expansion of a quiescent Treg precursor population, which is CD4+CD25-Foxp3+.


Asunto(s)
Antineoplásicos/metabolismo , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Recuento de Células , Línea Celular Tumoral , Proliferación Celular , Femenino , Homeostasis , Interferón gamma/metabolismo , Ganglios Linfáticos/citología , Ratones Noqueados , Neoplasias/patología , Bazo/citología , Linfocitos T Reguladores/inmunología , Carga Tumoral
2.
BMC Cancer ; 16: 199, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26957307

RESUMEN

BACKGROUND: Alpha-tocopheryloxyacetic acid (α-TEA) is a semi-synthetic derivative of naturally occurring vitamin E (alpha-tocopherol) that can be delivered via an oral route. Preclinical in vitro and in vivo data demonstrated that α-TEA is a potent anti-tumor agent with a safe toxicity profile in mice. We report a comprehensive study to evaluate the toxokinetics of good manufacturing practice (GMP)-grade α-TEA in dogs after daily oral administration for 28 days, followed by a 28-day recovery period. METHODS: Male and female beagle dogs received capsules of α-TEA Lysine Salt at doses of 100, 300, 1500 mg/kg/day. α-TEA plasma levels were determined by high-performance liquid chromatography (HPLC) with mass spectrometric detection. During the treatment, animals were observe for clinical signs, food consumption, body weight, and subjected to ophthalmoscopic, and electrocardiographic assessments. At the end of the dosing period, blood was taken and toxicokinetic analyses and histopathology assessments were performed when animals were necropsied. RESULTS: Our findings showed that there was no α-TEA-related mortality or moribundity. At the highest dose, increases in white blood cells and fibrinogen levels were observed. These levels returned to normal at the end of the recovery period. Histopathological evaluation of major organs revealed no significant lesions related to α-TEA-treatment. CONCLUSION: We demonstrate that for designing clinical trials in patients, the highest non-severely toxic dose (HNSTD) of α-TEA is 1500 mg/kg/day in Beagle dogs and this data informed the design of dose-escalation studies of α-TEA in patients with advanced cancer.


Asunto(s)
Tocoferoles/farmacocinética , Tocoferoles/toxicidad , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Biopsia , Recuento de Células Sanguíneas , Análisis Químico de la Sangre , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea , Perros , Femenino , Lisina , Masculino , Sales (Química) , Factores de Tiempo , Tocoferoles/administración & dosificación , Tocoferoles/química , Pruebas de Toxicidad , Toxicocinética , Urinálisis
3.
Biochim Biophys Acta ; 1817(9): 1597-607, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22627082

RESUMEN

The effects of α-tocopheryl succinate (α-TOS), α-tocopheryl acetyl ether (α-TEA) and triphenylphosphonium-tagged vitamin E succinate (mitochondrially targeted vitamin E succinate; MitoVES) on energy-related mitochondrial functions were determined in mitochondria isolated from AS-30D hepatoma and rat liver, bovine heart sub-mitochondrial particles (SMPs), and in rodent and human carcinoma cell lines and rat hepatocytes. In isolated mitochondria, MitoVES stimulated basal respiration and ATP hydrolysis, but inhibited net state 3 (ADP-stimulated) respiration and Ca(2+) uptake, by collapsing the membrane potential at low doses (1-10µM). Uncoupled mitochondrial respiration and basal respiration of SMPs were inhibited by the three drugs at concentrations at least one order of magnitude higher and with different efficacy: MitoVES>α-TEA>α-TOS. At high doses (>10µM), the respiratory complex II (CII) was the most sensitive MitoVES target. Acting as an uncoupler at low doses, this agent stimulated total O(2) uptake, collapsed ∆ψ(m), inhibited oxidative phosphorylation and induced ATP depletion in rodent and human cancer cells more potently than in normal rat hepatocytes. These findings revealed that in situ tumor mitochondria are preferred targets of the drug, indicating its clinical relevance.


Asunto(s)
Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Vitamina E/análogos & derivados , Adenosina Trifosfato/química , Animales , Calcio/metabolismo , Bovinos , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/fisiología , Ratas
4.
Front Immunol ; 14: 1057702, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911733

RESUMEN

Cancer immunotherapy such as anti-PD-1/anti-PD-L1 immune checkpoint blockade (ICB) can provide significant clinical benefit in patients with advanced malignancies. However, most patients eventually develop progressive disease, thus necessitating additional therapeutic options. We have developed a novel agent, a-TEA-LS, that selectively induces tumor cell death while sparing healthy tissues, leading to increased activation of tumor-reactive T cells and tumor regression. In the current study, we explored the impact of combined a-TEA-LS + ICB in orthotopic and spontaneously arising murine models of mammary carcinoma. We found that a-TEA-LS + ICB led to increased production of pro-inflammatory cytokines that were associated with a reduction in tumor growth and prolonged survival. Together, these data demonstrate the potential utility of a-TEA-LS + ICB for the treatment of breast cancer and provide the rationale for clinical translation of this novel approach.


Asunto(s)
Neoplasias de la Mama , Inhibidores de Puntos de Control Inmunológico , Humanos , Animales , Ratones , Femenino , Inmunoterapia , Citocinas
5.
Int J Cancer ; 131(5): 1052-8, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22038845

RESUMEN

Breast cancer is the number one neoplastic disease of women, with the HER2-high carcinomas presenting a considerable challenge for efficient treatment. Therefore, a search for novel agents active against this type of cancer is warranted. We tested two vitamin E (VE) analogs, the esterase-hydrolyzable α-tocopheryl succinate (α-TOS) and the non-hydrolyzable ether α-tocopheryloxyacetic acid (α-TEA) for their effects on HER2-positive breast carcinomas using a breast tumor mouse model and breast cancer cell lines. Ultrasound imaging documented that α-TEA suppressed breast carcinomas in the transgenic animals more efficiently than found for its ester counterpart. However, both agents exerted a comparable apoptotic effect on the NeuTL breast cancer cells derived from the FVB/N c-neu mice as well as in the human MBA-MD-453 and MCF7HER2-18 cells with high level of HER2. The superior anti-tumor effect of α-TEA over α-TOS in vivo can be explained by longer persistence of the former in mice, possibly due to the enhanced plasma and hepatic processing of α-TOS in comparison to the esterase-non-cleavable α-TEA. Indeed, the stability of α-TOS in plasma was inferior to that of α-TEA. We propose that α-TEA is a promising drug efficient against breast cancer, as documented by its effect on experimental HER2-positive breast carcinomas that present a considerable problem in cancer management.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Receptor ErbB-2/metabolismo , Tocoferoles/química , Tocoferoles/uso terapéutico , alfa-Tocoferol/química , alfa-Tocoferol/uso terapéutico , Animales , Antioxidantes/química , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
6.
Cancer Immunol Immunother ; 61(4): 511-21, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21971588

RESUMEN

Effective tumor immunotherapy may require not only activation of anti-tumor effector cells, but also abrogation of tumor-mediated immunosuppression. The cytokine TGF-ß, is frequently elevated in the tumor microenvironment and is a potent immunosuppressive agent and promoter of tumor metastasis. OX40 (CD134) is a member of the TNF-α receptor superfamily and ligation by agonistic antibody (anti-OX40) enhances effector function, expansion, and survival of activated T cells. In this study, we examined the therapeutic efficacy and anti-tumor immune response induced by the combination of a small molecule TGF-ß signaling inhibitor, SM16, plus anti-OX40 in the poorly immunogenic, highly metastatic, TGF-ß-secreting 4T1 mammary tumor model. Our data show that SM16 and anti-OX40 mutually enhanced each other to elicit a potent anti-tumor effect against established primary tumors, with a 79% reduction in tumor size, a 95% reduction in the number of metastatic lung nodules, and a cure rate of 38%. This positive treatment outcome was associated with a 3.2-fold increase of tumor-infiltrating, activated CD8+ T cells, an overall accumulation of CD4+ and CD8+ T cells, and an increased tumor-specific effector T cell response. Complete abrogation of the therapeutic effect in vivo following depletion of CD4+ and CD8+ T cells suggests that the anti-tumor efficacy of SM16+ anti-OX40 therapy is T cell dependent. Mice that were cured of their tumors were able to reject tumor re-challenge and manifested a significant tumor-specific peripheral memory IFN-γ response. Taken together, these data suggest that combining a TGF-ß signaling inhibitor with anti-OX40 is a viable approach for treating metastatic breast cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Compuestos de Azabiciclo/administración & dosificación , Carcinoma/tratamiento farmacológico , Inmunoterapia , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Compuestos de Azabiciclo/efectos adversos , Carcinoma/patología , Progresión de la Enfermedad , Sinergismo Farmacológico , Femenino , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Trasplante de Neoplasias , Receptores OX40/agonistas , Receptores OX40/inmunología , Transducción de Señal/efectos de los fármacos , Carga Tumoral
7.
Anticancer Drugs ; 23(4): 455-64, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22185820

RESUMEN

Alpha-tocopheryloxy acetic acid (α-TEA) is an ether derivative of vitamin E and has been shown to suppress tumor growth in various murine and human xenograft tumor models, including melanoma, breast, lung, prostate, and ovarian cancers. The purpose of this study was to assess its safety and pharmacokinetics after repeat dosing in a preclinical murine model. Male and female mice received α-TEA doses of 100, 300, or 1500 mg/kg/day by daily oral gavage for 28 days. α-TEA serum levels were determined weekly by high-performance liquid chromatography with mass spectrometric detection. After 28 days of dosing, complete blood counts were taken, blood chemistry was analyzed, and histology was performed. Pharmacokinetic parameters were determined after single dosing. There was no mortality, and we found no clinical signs of toxicity in any of the α-TEA doses tested. Histopathological evaluation of major organs (heart, lung, kidney, liver, spleen, jejunum, ileum, and cecum) revealed no significant α-TEA treatment-related lesions. Blood counts revealed low-grade anemia but no other significant differences between treatment and control groups. Blood chemistry revealed moderate liver toxicity that was dose dependent and was absent in the lowest dose group. There were no significant sex-specific differences in the toxicity profile. The half-life of orally administered α-TEA was determined to be 52 h. This is the first report comprehensively evaluating the toxicity profile of this novel anticancer drug and will facilitate the design of clinical trials to evaluate the safety and antitumor efficacy of α-TEA in patients with cancer.


Asunto(s)
Antineoplásicos/farmacocinética , Tocoferoles/farmacocinética , Anemia/inducido químicamente , Animales , Antineoplásicos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Tocoferoles/toxicidad
8.
Breast Cancer Res ; 13(1): R4, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21232138

RESUMEN

INTRODUCTION: α-Tocopheryloxyacetic acid (α-TEA) is a novel ether derivative of α-tocopherol that has generated interest as a chemotherapeutic agent because of its selective toxicity toward tumor cells and its ability to suppress tumor growth in various rodent and human xenograft models. We previously reported that oral α-TEA inhibited the growth of both a transplanted (4T1) and a spontaneous MMTV-PyMT mouse model of breast cancer. METHODS: Because little is known about the possible immunological mechanisms underlying the in vivo α-TEA effects, we evaluated the impact of α-TEA therapy on the immune response by characterizing immune cell populations infiltrating the tumor site. RESULTS: α-TEA treatment resulted in higher frequencies of activated T cells in the tumor microenvironment and twofold and sixfold higher ratios of CD4⁺ and CD8⁺T cells to regulatory T cells, respectively. This finding was correlated with an increased ability of tumor-draining lymph node cells and splenocytes from α-TEA-treated mice to secrete interferon (IFN)-γ in response to CD3 or to mediate a cytolytic response in a tumor-specific fashion, respectively. That the α-TEA-mediated antitumor effect had a T cell-dependent component was demonstrated by the partial abrogation of tumor suppression when CD4⁺ and CD8⁺ T cells were depleted. We also determined the intratumoral cytokine and chemokine profile and found that α-TEA treatment increased intratumoral IFN-γ levels but decreased interleukin (IL)-4 levels, suggesting a shift toward a TH1 response. In addition, α-TEA induced higher levels of the inflammatory cytokine IL-6 and the chemokine CCL5. CONCLUSIONS: Taken together, these data suggest that α-TEA treatment, in addition to its direct cytotoxic effects, enhanced the anti-tumor immune response. This study provides a better understanding of the mechanisms of action of α-TEA and its effect on the immune system and may prove useful in designing immune-stimulating strategies to boost the antitumor effects of α-TEA in breast cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Mamarias Experimentales/inmunología , Tocoferoles/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Estimación de Kaplan-Meier , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Tocoferoles/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
9.
BMC Cancer ; 11: 471, 2011 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22044845

RESUMEN

BACKGROUND: HER2/neu is an oncogene that facilitates neoplastic transformation due to its ability to transduce growth signals in a ligand-independent manner, is over-expressed in 20-30% of human breast cancers correlating with aggressive disease and has been successfully targeted with trastuzumab (Herceptin®). Because trastuzumab alone achieves only a 15-30% response rate, it is now commonly combined with conventional chemotherapeutic drugs. While the combination of trastuzumab plus chemotherapy has greatly improved response rates and increased survival, these conventional chemotherapy drugs are frequently associated with gastrointestinal and cardiac toxicity, bone marrow and immune suppression. These drawbacks necessitate the development of new, less toxic drugs that can be combined with trastuzumab. Recently, we reported that orally administered alpha-tocopheryloxyacetic acid (α-TEA), a novel ether derivative of alpha-tocopherol, dramatically suppressed primary tumor growth and reduced the incidence of lung metastases both in a transplanted and a spontaneous mouse model of breast cancer without discernable toxicity. METHODS: In this study we examined the effect of α-TEA plus HER2/neu-specific antibody treatment on HER2/neu-expressing breast cancer cells in vitro and in a HER2/neu positive human xenograft tumor model in vivo. RESULTS: We show in vitro that α-TEA plus anti-HER2/neu antibody has an increased cytotoxic effect against murine mammary tumor cells and human breast cancer cells and that the anti-tumor effect of α-TEA is independent of HER2/neu status. More importantly, in a human breast cancer xenograft model, the combination of α-TEA plus trastuzumab resulted in faster tumor regression and more tumor-free animals than trastuzumab alone. CONCLUSION: Due to the cancer cell selectivity of α-TEA, and because α-TEA kills both HER2/neu positive and HER2/neu negative breast cancer cells, it has the potential to be effective and less toxic than existing chemotherapeutic drugs when used in combination with HER2/neu antibody.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Antioxidantes/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Tocoferoles/farmacología , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Humanos , Ratones , Modelos Biológicos , Fragmentos de Péptidos/metabolismo , Trastuzumab
10.
Anticancer Res ; 29(6): 2099-109, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19528470

RESUMEN

BACKGROUND: Transforming growth factor beta (TGF-beta) plays a complex role in breast carcinogenesis. Initially functioning as a tumor suppressor, this cytokine later contributes to the progression of malignant cells by enhancing their invasive and metastatic potential as well as suppressing antitumor immunity. The purpose of this study was to investigate the efficacy of SM16, a novel small molecule ALK5 kinase inhibitor, to treat a highly metastatic, TGF-beta-producing murine mammary carcinoma (4T1). MATERIALS AND METHODS: Mice bearing established 4T1 tumors were treated with SM16 intraperitoneally (i.p.) or orally, and primary and metastatic tumor growth was assessed. RESULTS: SM16 inhibited Smad2 phosphorylation in cultured 4T1 tumor cells as well as primary and metastatic 4T1 tumor tissue. Blockade of TGF-beta signal transduction in 4T1 tumor cells by SM16 prevented TGF-beta-induced morphological changes and inhibited TGF-beta-induced invasion in vitro. When delivered via daily i.p. injection or orally through mouse chow, SM16 inhibited the growth of primary and metastatic 4T1 tumors. Splenocytes isolated from mice on the SM16 diet displayed enhanced IFN-gamma production and antitumor CTL activity. Furthermore, SM16 failed to inhibit the growth and metastasis of established 4T1 tumors in immunodeficient SCID mice. CONCLUSION: Taken together, the data indicate that the antitumor efficacy of SM16 is dependent on an immune-mediated mechanism and that SM16 may represent a safe and effective treatment for metastatic breast cancer.


Asunto(s)
Compuestos de Azabiciclo/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Front Oncol ; 9: 1554, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32039024

RESUMEN

Tumors employ strategies to escape immune control. The principle aim of most cancer immunotherapies is to restore effective immune surveillance. Among the different processes regulating immune escape, tumor microenvironment-associated soluble factors, and/or cell surface-bound molecules are mostly responsible for dysfunctional activity of tumor-specific CD8+T cells. These dynamic immunosuppressive networks prevent tumor rejection at several levels, limiting also the success of immunotherapies. Nevertheless, the recent clinical development of immune checkpoint inhibitors or of molecules modulating cellular targets and immunosuppressive enzymes highlights the great potential of approaches based on the selective disruption of immunosuppressive networks. Currently, the administration of different categories of immunotherapy in combination regimens is the ultimate modality for impacting the survival of cancer patients. With the advent of immune checkpoint inhibitors, designed to mount an effective antitumor immune response, profound changes occurred in cancer immunotherapy: from a global stimulation of the immune system to a specific targeting of an immune component. This review will specifically highlight the players, the mechanisms limiting an efficient antitumor response and the current immunotherapy modalities tailored to target immune suppressive pathways. We also discuss the ongoing challenges encountered by these strategies and provide suggestions for circumventing hurdles to new immunotherapeutic approaches, including the use of relevant biomarkers in the optimization of immunotherapy regimens and the identification of patients who can benefit from defined immune-based approaches.

12.
Mol Aspects Med ; 28(5-6): 607-45, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17499351

RESUMEN

Mitochondria have recently emerged as new and promising targets for cancer prevention and therapy. One of the reasons for this is that mitochondria are instrumental to many types of cell death and often lie downstream from the initial actions of anti-cancer drugs. Unlike the tumour suppressor gene encoding p53 that is notoriously prone to inactivating mutations but whose function is essential for induction of apoptosis by DNA-targeting agents (such as doxorubicin or 5-fluorouracil), mitochondria present targets that are not so compromised by genetic mutation and whose targeting overcomes problems with mutations of upstream targets such as p53. We have recently proposed a novel class of anti-cancer agents, mitocans that exert their anti-cancer activity by destabilising mitochondria, promoting the selective induction of apoptotic death in tumour cells. In this communication, we review recent findings on mitocans and propose a common basis for their mode of action in inducing apoptosis of cancer cells. We use as an example the analogues of vitamin E that are proving to be cancer cell-specific and may soon be developed into efficient anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Mitocondrias/efectos de los fármacos , Vitamina E/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
13.
Cancer Res ; 64(21): 8085-92, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520220

RESUMEN

In this study, we show that rodent albumin is expressed by and cell surface localized on at least some murine tumor cells. We have been able to purify this tumor-expressed albumin from in vivo grown tumor masses. The tumor-expressed albumin, unlike normal serum albumin purified from blood, is capable of inhibiting T-cell activation, proliferation, and function in both in vitro and in vivo settings. Tumor-expressed albumin does not appear to affect antigen processing or presentation by professional antigen-presenting cells. The activity appears to lie in relatively small, lipid-like moieties that are presumably cargo for tumor-expressed albumin, and that activity can be removed from the albumin by lipid removal or treatment with lipase. Thus, we herein report of a novel form of tumor-induced immune suppression attributable to lipid-like entities, cloaked by albumin produced by tumors.


Asunto(s)
Albúminas/farmacología , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Proteínas de Neoplasias/farmacología , Linfocitos T/inmunología , Albúminas/química , Albúminas/aislamiento & purificación , Animales , Lípidos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/inmunología
14.
Cancer Res ; 63(8): 1860-4, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12702574

RESUMEN

Dendritic cell (DC)-based vaccines have exhibited minimal effectiveness in treating established tumors, likely because of factors present in the tumor microenvironment. One such factor is transforming growth factor beta (TGF-beta), a cytokine that is produced by numerous tumor types and has been demonstrated to impair DC functions in vitro. We have evaluated the effect of TGF-beta on the immunostimulatory activities of DCs. We demonstrate that TGF-beta exposure inhibits the ability of DCs to present antigen, stimulate tumor-sensitized T lymphocytes, and migrate to draining lymph nodes. Neutralization of TGF-beta using the TGF-beta-neutralizing monoclonal antibody 2G7 enhanced the ability of DC vaccines to inhibit the growth of established 4T1 murine mammary tumors. Treatment of 4T1 tumors transduced with the antisense TGF-beta transgene (4T1-asT) with the combination of DC and 2G7 monoclonal antibody inhibited tumor growth and resulted in complete regression of tumors in 40% of the mice. These results demonstrate that neutralization of TGF-beta in tumor-bearing mice enhances the efficacy of DC-based vaccines.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Presentación de Antígeno/efectos de los fármacos , Vacunas contra el Cáncer/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Factor de Crecimiento Transformador beta/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Presentación de Antígeno/inmunología , Antígenos CD40/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , ADN sin Sentido/genética , ADN sin Sentido/farmacología , Femenino , Inmunoterapia Adoptiva , Ganglios Linfáticos/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
15.
Oncotarget ; 7(48): 78653-78666, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27602953

RESUMEN

Radiation therapy provides a means to kill large numbers of cancer cells in a controlled location resulting in the release of tumor-specific antigens and endogenous adjuvants. However, by activating pathways involved in apoptotic cell recognition and phagocytosis, irradiated cancer cells engender suppressive phenotypes in macrophages. We demonstrate that the macrophage-specific phagocytic receptor, Mertk is upregulated in macrophages in the tumor following radiation therapy. Ligation of Mertk on macrophages results in anti-inflammatory cytokine responses via NF-kB p50 upregulation, which in turn limits tumor control following radiation therapy. We demonstrate that in immunogenic tumors, loss of Mertk is sufficient to permit tumor cure following radiation therapy. However, in poorly immunogenic tumors, TGFß inhibition is also required to result in tumor cure following radiation therapy. These data demonstrate that Mertk is a highly specific target whose absence permits tumor control in combination with radiation therapy.


Asunto(s)
Macrófagos/efectos de la radiación , Recurrencia Local de Neoplasia , Neoplasias Experimentales/radioterapia , Tirosina Quinasa c-Mer/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Compuestos de Azabiciclo/farmacología , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Células RAW 264.7 , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de la radiación , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Tirosina Quinasa c-Mer/antagonistas & inhibidores , Tirosina Quinasa c-Mer/deficiencia , Tirosina Quinasa c-Mer/genética
16.
Cancer Immunol Res ; 3(5): 526-35, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25627655

RESUMEN

In preclinical tumor models, αOX40 therapy is often successful at treating small tumors, but is less effective once the tumors become large. For a tumor immunotherapy to be successful to cure large tumors, it will most likely require not only an agonist to boost effector T-cell function but also inhibitors of T-cell suppression. In this study, we show that combining αOX40 antibodies with an inhibitor of the TGFß receptor (SM16) synergizes to elicit complete regression of large established MCA205 and CT26 tumors. Evaluation of tumor-infiltrating T cells showed that SM16/αOX40 dual therapy resulted in an increase in proliferating granzyme B(+) CD8 T cells, which produced higher levels of IFNγ, compared with treatment with either agent alone. We also found that the dual treatment increased pSTAT3 expression in both CD4 and CD8 T cells isolated from tumors. Because others have published that STAT3 signaling is detrimental to T-cell function within the tumor microenvironment, we explored whether deletion of STAT3 in OX40-expressing cells would affect this potent combination therapy. Surprisingly, we found that deletion of STAT3 in OX40-expressing cells decreased the efficacy of this combination therapy, showing that the full therapeutic potential of this treatment depends on STAT3 signaling, most likely in the T cells of tumor-bearing mice.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias/metabolismo , Receptores OX40/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Compuestos de Azabiciclo/administración & dosificación , Línea Celular Tumoral , Femenino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptores OX40/inmunología , Transducción de Señal
17.
Cancer Immunol Res ; 2(10): 1011-22, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047233

RESUMEN

The immune infiltrate in colorectal cancer has been correlated with outcome, such that individuals with higher infiltrations of T cells have increased survival independent of the disease stage. For patients with lower immune infiltrates, overall survival is limited. Because the patients with colorectal cancer studied have received conventional cancer therapies, these data may indicate that the pretreatment tumor environment increases the efficacy of treatments such as chemotherapy, surgery, and radiotherapy. This study was designed to test the hypothesis that an improved immune environment in the tumor at the time of treatment will increase the efficacy of radiotherapy. We demonstrate that inhibition of TGFß using the orally available small-molecule inhibitor SM16 improved the immune environment of tumors in mice and significantly improved the efficacy of subsequent radiotherapy. This effect was not due to changes in radiosensitivity, epithelial-mesenchymal transition, or changes in vascular function in the tumor; rather, this effect was dependent on adaptive immunity and resulted in long-term protective immunity in cured mice. These data demonstrate that immunotherapy is an option to improve the immune status of patients with poor tumor infiltrates and that pretreatment improves the efficacy of radiotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos de Azabiciclo/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/radioterapia , Evaluación Preclínica de Medicamentos/métodos , Femenino , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Neoadyuvante/métodos , Trasplante de Neoplasias , Tolerancia a Radiación/efectos de los fármacos , Células Tumorales Cultivadas
18.
Autophagy ; 9(3): 429-31, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23235369

RESUMEN

Stimulation of apoptosis has been reported as the primary mechanism of tumor cell death induced by alpha-tocopheryloxyacetic acid (α-TEA), an esterase-resistant, semi-synthetic derivative of vitamin E (R-R-R-α-tocopherol). New information now shows that α-TEA also triggers tumor cell autophagy and promotes antigen cross-presentation. Autophagosome-enriched fractions of α-TEA-treated tumor cells (α-TAGS) efficiently cross-primed antigen-specific CD8 (+) T cells and vaccination with dendritic cells (DC) pulsed with α-TAGS reduced lung metastases and increased survival of tumor-bearing mice. Taken together, these observations suggest that both autophagy and apoptosis signaling programs are activated in tumor cells by α-TEA treatment and may contribute to tumor cell death. We propose that autophagy-dependent enhancement of cross-presentation is a novel mechanism of α-TEA-mediated tumor immunity and that α-TEA can be exploited as an adjuvant to enhance the antitumor response.


Asunto(s)
Presentación de Antígeno , Apoptosis , Autofagia/fisiología , Regulación Neoplásica de la Expresión Génica , Tocoferoles/farmacología , Animales , Autofagia/efectos de los fármacos , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Supervivencia Celular , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Tocoferoles/química , Vitamina E/análogos & derivados
19.
Cancer Res ; 72(14): 3535-45, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22745370

RESUMEN

The semisynthetic vitamin E derivative alpha-tocopheryloxyacetic acid (α-TEA) induces tumor cell apoptosis and may offer a simple adjuvant supplement for cancer therapy if its mechanisms can be better understood. Here we report that α-TEA also triggers tumor cell autophagy and that it improves cross-presentation of tumor antigens to the immune system. α-TEA stimulated both apoptosis and autophagy in murine mammary and lung cancer cells and inhibition of caspase-dependent apoptosis enhanced α-TEA-induced autophagy. Cell exposure to α-TEA generated double-membrane-bound vesicles indicative of autophagosomes, which efficiently cross-primed antigen-specific CD8(+) T cells. Notably, vaccination with dendritic cells pulsed with α-TEA-generated autophagosomes reduced lung metastases and increased the survival of tumor-bearing mice. Taken together, our findings suggest that both autophagy and apoptosis signaling programs are activated during α-TEA-induced tumor cell killing. We suggest that the ability of α-TEA to stimulate autophagy and enhance cross-priming of CD8(+) T cells might be exploited as an adjuvant strategy to improve stimulation of antitumor immune responses.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Presentación de Antígeno/efectos de los fármacos , Autofagia/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Animales/tratamiento farmacológico , Tocoferoles/farmacología , Adenocarcinoma/inmunología , Adenocarcinoma del Pulmón , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Supervivencia Celular , Células Dendríticas/inmunología , Femenino , Neoplasias Pulmonares/inmunología , Activación de Linfocitos , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Vacunación
20.
Antioxid Redox Signal ; 15(12): 2989-3002, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21504364

RESUMEN

SIGNIFICANCE: Recent research has shown that tumors contain a small subpopulation of stem-like cells that are more resistant to therapy and that are likely to produce second-line tumors. RECENT ADVANCES: Cancer stem-like cells (CSCs) have been characterized by a variety of markers, including, for a number of types of cancer, high expression of the plasma membrane protein CD133, which is also indicative of the increase of stemness of cultured cancer cells growing as spheres. CRITICAL ISSUES: While the function of this protein has not yet been clearly defined, it may have a role in the stem-like phenotype of CSCs that cause (re-)initiation of tumors as well as their propagation. We hypothesize that CD133 selects for CSC survival against not only immunosurveillance mechanisms but also stress-induced apoptosis. FUTURE DIRECTIONS: High level of expression of CD133 may be a useful marker of more aggressive tumors that are recalcitrant toward established therapies. Compelling preliminary data indicate that drugs targeting mitochondria may be utilized as a novel, efficient cancer therapeutic modality.


Asunto(s)
Antígenos CD/metabolismo , Apoptosis , Glicoproteínas/metabolismo , Vigilancia Inmunológica , Mitocondrias/metabolismo , Péptidos/metabolismo , Antígeno AC133 , Animales , Antígenos CD/genética , Apoptosis/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Humanos , Vigilancia Inmunológica/genética , Células Madre Neoplásicas/metabolismo , Péptidos/genética , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA