Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 20(1): 603, 2020 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-32600429

RESUMEN

BACKGROUND: Modulated electro-hyperthermia (mEHT) is a form of hyperthermia used in cancer treatment. mEHT has demonstrated the ability to activate host immunity by inducing the release of heat shock proteins, triggering apoptosis, and destroying the integrity of cell membranes to enhance cellular uptake of chemo-drugs in tumor cells. Both curcumin and resveratrol are phytochemicals that function as effective antioxidants, immune activators, and potential inhibitors of tumor development. However, poor bioavailability is a major obstacle for use in clinical cancer treatment. METHODS: This purpose of this study was to investigate whether mEHT can increase anti-cancer efficacy of nanosized curcumin and resveratrol in in vitro and in vivo models. The in vitro study included cell proliferation assay, cell cycle, and apoptosis analysis. Serum concentration was analyzed for the absorption of curcumin and resveratrol in SD rat model. The in vivo CT26/BALB/c animal tumor model was used for validating the safety, tumor growth curve, and immune cell infiltration within tumor tissues after combined mEHT/curcumin/resveratrol treatment. RESULTS: The results indicate co-treatment of mEHT with nano-curcumin and resveratrol significantly induced cell cycle arrest and apoptosis of CT26 cells. The serum concentrations of curcumin and resveratrol were significantly elevated when mEHT was applied. The combination also inhibited the growth of CT26 colon cancer by inducing apoptosis and HSP70 expression of tumor cells while recruiting CD3+ T-cells and F4/80+ macrophages. CONCLUSIONS: The results of this study have suggested that this natural, non-toxic compound can be an effective anti-tumor strategy for clinical cancer therapy. mEHT can enable cellular uptake of potential anti-tumor materials and create a favorable tumor microenvironment for an immunological chain reaction that improves the success of combined treatments of curcumin and resveratrol.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias Colorrectales/terapia , Curcumina/administración & dosificación , Terapia por Estimulación Eléctrica/métodos , Hipertermia Inducida/métodos , Resveratrol/administración & dosificación , Animales , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Disponibilidad Biológica , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Línea Celular Tumoral/trasplante , Neoplasias Colorrectales/patología , Terapia Combinada/métodos , Curcumina/efectos adversos , Curcumina/farmacocinética , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Nanopartículas/administración & dosificación , Ratas , Resveratrol/efectos adversos , Resveratrol/farmacocinética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
2.
Theranostics ; 10(17): 7510-7526, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685002

RESUMEN

Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor immune responses (M1). Recent studies have shown that c-MYC (MYC) is induced in M2 macrophages in vitro and in vivo where it regulates the expression of tumor-promoting genes. In a myeloid lineage MYC KO mouse model, MYC had important roles in macrophage maturation and function leading to reduced tumor growth. We therefore hypothesized that targeted delivery of a MYC inhibitor to established M2 TAMs could reduce polarization toward an M2 phenotype in breast cancer models. Methods: In this study, we developed a MYC inhibitor prodrug (MI3-PD) for encapsulation within perfluorocarbon nanoparticles, which can deliver drugs directly to the cytosol of the target cell through a phagocytosis independent mechanism. We have previously shown that M2-like TAMs express significant levels of the vitronectin receptor, integrin ß3, and in vivo targeting and therapeutic potential was evaluated using αvß3 integrin targeted rhodamine-labeled nanoparticles (NP) or integrin αvß3-MI3-PD nanoparticles. Results: We observed that rhodamine, delivered by αvß3-rhodamine NP, was incorporated into M2 tumor promoting macrophages through both phagocytosis-independent and dependent mechanisms, while NP uptake in tumor suppressing M1 macrophages was almost exclusively through phagocytosis. In a mouse model of breast cancer (4T1-GFP-FL), M2-like TAMs were significantly reduced with αvß3-MI3-PD NP treatment. To validate this effect was independent of drug delivery to tumor cells and was specific to the MYC inhibitor, mice with integrin ß3 knock out tumors (PyMT-Bo1 ß3KO) were treated with αvß3-NP or αvß3-MI3-PD NP. M2 macrophages were significantly reduced with αvß3-MI3-PD nanoparticle therapy but not αvß3-NP treatment. Conclusion: These data suggest αvß3-NP-mediated drug delivery of a c-MYC inhibitor can reduce protumor M2-like macrophages while preserving antitumor M1-like macrophages in breast cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Nanopartículas/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Antineoplásicos/química , Neoplasias de la Mama/patología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Línea Celular Tumoral/trasplante , Evaluación Preclínica de Medicamentos , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/química , Técnicas de Inactivación de Genes , Humanos , Integrina alfaVbeta3 , Integrina beta3 , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Nanopartículas/química , Fagocitosis , Cultivo Primario de Células , Profármacos/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
3.
Mol Pharm ; 17(10): 3720-3729, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32633977

RESUMEN

The limited tumor tissue penetration of many nanoparticles remains a formidable challenge to their therapeutic efficacy. Although several photonanomedicines have been applied to improve tumor penetration, the first near-infrared window mediated by the low optical tissue penetration depth severely limits their anticancer effectiveness. To achieve deep optical tissue and drug delivery penetration, a near-infrared second window (NIR-II)-excited and pH-responsive ultrasmall drug delivery nanoplatform was fabricated based on BSA-stabilized CuS nanoparticles (BSA@CuS NPs). The BSA@CuS NPs effectively encapsulated doxorubicin (DOX) via strong electrostatic interactions to form multifunctional nanoparticles (BSA@CuS@DOX NPs). The BSA@CuS@DOX NPs had an ultrasmall size, which allowed them to achieve deeper tumor penetration. They also displayed stronger NIR II absorbance-mediated deep optical tissue penetration than that of the NIR I window. Moreover, the multifunctional nanoplatform preferentially accumulated in tumor sites, induced tumor hyperthermia, and generated remarkably high ROS levels in tumor sites upon NIR-II laser (1064 nm) irradiation. More importantly, our strategy achieved excellent synergistic effects of chemotherapy and phototherapy (chemophototherapy) under the guidance of photothermal imaging. The developed nanoparticles also showed good biocompatibility and bioclearance properties. Therefore, our work demonstrated a facile strategy for fabricating a multifunctional nanoplatform that is a promising candidate for deep tumor penetration as an effective antitumor therapy.


Asunto(s)
Doxorrubicina/administración & dosificación , Portadores de Fármacos/efectos de la radiación , Nanopartículas/efectos de la radiación , Neoplasias/tratamiento farmacológico , Fototerapia/métodos , Animales , Línea Celular Tumoral/trasplante , Supervivencia Celular , Modelos Animales de Enfermedad , Doxorrubicina/farmacocinética , Portadores de Fármacos/química , Liberación de Fármacos/efectos de la radiación , Humanos , Concentración de Iones de Hidrógeno , Rayos Infrarrojos , Rayos Láser , Ratones , Nanopartículas/química , Neoplasias/patología , Fototerapia/instrumentación , Distribución Tisular
4.
Sci Rep ; 10(1): 5925, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32245980

RESUMEN

The efficacy of different modalities of treating breast cancer is inhibited by several limitations such as off-targeted drug distribution, rapid drug clearance, and drug resistance. To overcome these limitations, we developed Lf-Doxo-PMNSs for combined chemo-MF-PTT. The PMNSs were synthesized by hydrothermal method and their physicochemical properties were examined by FE-SEM, TEM, DLS, TGA, XRD investigations. The cytotoxicity of as-synthesized NPs against 4T1 cells was carried out by MTT and flow cytometry assays. Afterwards, the anti-cancer activities of as-synthesized Lf-Doxo-PMNSs on the tumor status, drug distribution and apoptosis mechanism were evaluated. The anti-cancer assays showed that Lf-Doxo-PMNSs significantly suppressed the cancer cell proliferation and tumor weight by prolonging drug availability and potential drug loading in tumor cells; whereas they showed a minimum cytotoxicity against non-cancerous cells. Likewise, combined chemo-MF-PTT using Lf-Doxo-PMNSs displayed the highest anti-cancer activity followed by combined chemo-PTT and combined chemo-MF therapy based on altering the apoptosis mechanism. Therefore, these results showed that combined chemo-MF-PTT based on Lf-Doxo-PMNSs can be used as a promising therapeutic platform with potential targeted drug delivery and high loading capacity features as well as reducing cancer drug resistance.


Asunto(s)
Neoplasias de la Mama/terapia , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Hipertermia Inducida/métodos , Magnetoterapia/métodos , Fotoquimioterapia/métodos , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral/trasplante , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacocinética , Liberación de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Nanopartículas de Magnetita/química , Ratones , Nanosferas/química , Distribución Tisular
5.
Int J Pharm ; 581: 119239, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32194211

RESUMEN

We induced changes in the tumor microenvironment (TME) through the synergistic actions of two drugs used in breast cancer therapy. The anti-fibrotic drug silibinin (SLB) targets tumor-associated fibroblasts and exerts immune-mediated anti-cancer effects. IPI-549, an efficient and highly selective phosphoinositide-3-kinase-gamma (PI3Kγ) inhibitor, was applied to alter the balance of immunosuppressive cells by inhibiting PI3Kγ molecules; it also promotes anti-tumor immunity. We developed nanoparticle formulations to encapsulate both drugs into the targeting carrier aminoethyl anisamide-polyethylene glycol-polycaprolactone (AEAA-PEG-PCL) respectively. The drugs were intravenously delivered in mice and resulted in an increase in anti-tumor efficacy and apoptotic tumor tissue compared with either IPI-549 or SLB alone in 4T1 breast cancer cell-derived tumors. Furthermore, a significant reduction in regulatory T (Treg) cells and myeloid suppressor cells (MDSCs) was observed. A normalized TME structure was also observed, including angiogenesis suppression, antifibrotic effects and the inhibition of collagen formation in the tumor tissue, significantly enhancing the anti-tumor effects. In summary, this combination strategy may offer an alternative treatment for breast cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Portadores de Fármacos/química , Isoquinolinas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Silibina/administración & dosificación , Administración Intravenosa , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Benzamidas/química , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Humanos , Isoquinolinas/farmacocinética , Ratones , Nanopartículas/química , Poliésteres/química , Polietilenglicoles/química , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Pirimidinas/farmacocinética , Silibina/farmacocinética , Microambiente Tumoral/efectos de los fármacos
6.
Int J Cancer ; 146(5): 1409-1420, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31702822

RESUMEN

Therapeutic success of targeted therapy with BRAF inhibitors (BRAFi) for melanoma is limited by resistance development. Observations from preclinical mouse models and recent insights into the immunological effects caused by BRAFi give promise for future development of combination therapy for human melanoma. In our study, we used the transplantable D4M melanoma mouse model with the BRAFV600E mutation and concomitant PTEN loss in order to characterize alterations in tumor-infiltrating effector immune cells when tumors become resistant to BRAFi. We found that BRAFi-sensitive tumors displayed a pronounced inflammatory milieu characterized by high levels of cytokines and chemokines accompanied by an infiltration of T and NK cells. The tumor-infiltrating effector cells were activated and produced high levels of IFN-γ, TNF-α and granzyme B. When tumors became resistant and progressively grew, they reverted to a low immunogenic state similar to untreated tumors as reflected by low mRNA levels of proinflammatory cytokines and chemokines and fewer tumor-infiltrating T and NK cells. Moreover, these T and NK cells were functionally impaired in comparison to their counterparts in BRAFi-sensitive tumors. Their effector cell function could be restored by additional peritumoral treatment with the TLR7 agonist imiquimod, a clinically approved agent for nonmelanoma skin cancer. Indeed, resistance to BRAFi therapy was delayed and accompanied by high numbers of activated T and NK cells in tumors. Thus, combining BRAFi with an immune stimulating agent such as a TLR ligand could be a promising alternative approach for the treatment of melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral/trasplante , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Humanos , Imiquimod/farmacología , Imiquimod/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Ratones , Mutación , Células T Asesinas Naturales , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo
7.
J Ethnopharmacol ; 249: 112412, 2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31751649

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Xiaoshui decoction (XSD) is a traditional Chinese medicine compound prescription that has been shown to reinforce the spleen and remove the fluid retention, while being widely used in the treatment of malignant pleural effusion (MPE). We previously reported that XSD alleviates symptoms and improves the quality of life in patients with MPE; however, the mechanism employed by XSD on MPE has not yet been elucidated. AIM OF THE STUDY: To investigate the role and mechanism of XSD in inhibiting the development of MPE, and in regulating macrophage polarization in vitro and in vivo. MATERIALS AND METHODS: A murine MPE model was used to study the effect of XSD on MPE. Mice with MPE were randomly allocated to a control group and XSD-low-dose (1.144 g/mL), XSD-middle-dose (2.288 g/mL), XSD-high-dose (4.576 g/mL), or cisplatin groups. RAW264.7 cells were induced to form tumor-associated macrophages (TAMs) as well as M1 and M2 macrophages using different conditioned media in vitro. RESULTS: XSD effectively inhibited MPE formation, reduced pleural permeability and angiogenesis, and prolonged mice survival. Particularly, XSD treatment induced the polarization of TAMs to the M1 phenotype in MPE. Moreover, in-vitro XSD remarkably promoted the expansion of M1 macrophages and reduced M2 macrophages by enhancing autophagy. CONCLUSIONS: XSD inhibits MPE development and regulates macrophage polarization by activating autophagy, indicating that XSD may serve as a novel option for integrative MPE therapies.


Asunto(s)
Autofagia/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Macrófagos/efectos de los fármacos , Derrame Pleural Maligno/tratamiento farmacológico , Animales , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Medicina Tradicional China/métodos , Ratones , Derrame Pleural Maligno/inmunología , Derrame Pleural Maligno/patología
8.
J Ethnopharmacol ; 247: 112256, 2020 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-31586690

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The mushroom Ganoderma lucidum (G. lucidum) is a traditional Chinese medicine reported to have a variety of pharmacological properties, including anti-cancer activity. G. lucidum spore oil (GLSO) is a lipid substance extracted from sporoderm-broken spore of G. lucidum. However, the effect of GLSO on breast cancer and the underlying molecular mechanism remain unclear. AIM OF THE STUDY: The aim of this study was to identify the effects of GLSO on breast cancer cells in vitro and in vivo as well as to investigate the mechanistic basis for the anticancer effect of GLSO. MATERIALS AND METHODS: First, in vitro MDA-MB-231 cells were treated with GLSO (0.2, 0.4, and 0.6 µL/mL). The protein levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), X-linked inhibitor of apoptosis (XIAP), total poly (ADP-ribose) polymerase (PARP), caspase-3 and caspase-8 were examined using western blotting. The mRNA expression levels of Fas-associated protein with death domain (FADD), TNF receptor-associated factor 2 (TRAF2), caspases-3, -8, -9 and Bax were examined using qRT-PCR. Second, in vivo the anticancer properties of GLSO were assessed by H&E, TUNEL and immunohistochemistry in BALB/c mice injected with 4T1 cells. In addition, the levels of caspase-9/caspase-3 signaling pathway proteins in tumor tissue were evaluated by immunoblotting. Finally, MDA-MB-231 cells were treated with caspase inhibitors to measure cell viability, the protein levels were examined with western blotting. RESULTS: The results in vitro showed that GLSO up-regulated the expression of Bax and caspase-3 in MDA-MB-231 cells, but had no effect on the expression of caspase-8. Moreover, the growth of tumors in vivo was significantly suppressed in the GLSO-treated group. The results of Western blot were consistent with in vitro. In vitro, co-treatment of MDA-MB-231 cells with caspase inhibitors reduced the inhibitory effect of GLSO on cell growth. CONCLUSIONS: GLSO inhibits the growth of MDA-MB-231 cells and tumors in vivo by inducing apoptosis, which may be achieved through the mitochondrial apoptotic pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Medicina Tradicional China/métodos , Aceites/farmacología , Reishi/química , Animales , Neoplasias de la Mama/patología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral/trasplante , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Aceites/uso terapéutico , Esporas Fúngicas/química
9.
Cancer Sci ; 110(9): 2933-2940, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31278877

RESUMEN

Chemotherapy has been the treatment of choice for unresectable peritoneal dissemination; however, it is difficult to eradicate such tumors because of poor drug delivery. To solve this issue, we developed FF-10832 as liposome-encapsulated gemcitabine to maintain a high concentration of gemcitabine in peritoneal tumors from the circulation and ascites. A syngeneic mouse model of peritoneal dissemination using murine Colon26 cell line was selected to compare the drug efficacy and pharmacokinetics of FF-10832 with those of gemcitabine. Despite the single intravenous administration, FF-10832 treatment enabled long-term survival of the lethal model mice as compared with those treated with gemcitabine. Pharmacokinetic analysis clarified that FF-10832 could achieve a more effective gemcitabine delivery to peritoneal tumors owing to better stability in the circulation and ascites. The novel liposome-encapsulated gemcitabine FF-10832 may be a curative therapeutic tool for cancer patients with unresectable peritoneal dissemination via the effective delivery of gemcitabine to target tumors.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Ascitis/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias Peritoneales/tratamiento farmacológico , Peritoneo/patología , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Ascitis/etiología , Línea Celular Tumoral/trasplante , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacocinética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Femenino , Humanos , Inyecciones Intravenosas , Estimación de Kaplan-Meier , Liposomas , Ratones , Ratones Endogámicos BALB C , Neoplasias Peritoneales/complicaciones , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/patología , Distribución Tisular , Resultado del Tratamiento , Gemcitabina
10.
Mol Pharm ; 16(8): 3430-3440, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31199661

RESUMEN

Poor anticancer ability, serious adverse reaction, and drug resistance against paclitaxel (PTX) have limited its clinical applications. When a mouse breast carcinoma cell line (4T1) was treated with both PTX and capsaicin (CAP), there was a synergistic anti-proliferative effect demonstrated with a combination index of 0.28. Therefore, a novel polyethylene glycol-derivatized CAP (PEG-Fmoc-CAP2) polymeric prodrug micellar carrier was developed and further encapsulated with PTX for antitumor combination treatment. The PEG-Fmoc-CAP2 polymeric micelles co-delivered with PTX achieved a 62.3% fraction of apoptotic cells in comparison to 45.4% fraction of apoptotic cells to that upon treatment with PTX alone. Comparable CAP amounts were found in the cell lysate treatment with PEG-Fmoc-CAP2-conjugated micelles to that of free CAP-treated 4T1 cells after 12 h treatment. Pharmacokinetic and biodistribution studies showed that the micelles possessed much longer circulation time in blood and preferential tumor tissue accumulation compared to the Taxol solution. Importantly, PTX/CAP-loaded micelles exhibited superior in vivo antitumor activity on the inhibition rate of tumor growth than other treatments (70.5% tumor growth reduction in PTX/CAP micelle-treated mice vs 57.8, 43.3, and 23.8% of tumor growth inhibition rate in PTX/PEG-Fmoc-OA2 micelles, Taxol, and PEG-Fmoc-CAP2 micelle-treated mice, respectively). Thus, the dual-functional PEG-Fmoc-CAP2 polymeric prodrug micelles are a promising co-delivery nanosystem for achieving synergistic antitumor efficacy of PTX and CAP.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Capsaicina/farmacología , Portadores de Fármacos/farmacología , Paclitaxel/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Capsaicina/química , Capsaicina/uso terapéutico , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Portadores de Fármacos/uso terapéutico , Sinergismo Farmacológico , Femenino , Humanos , Concentración 50 Inhibidora , Ratones , Micelas , Nanopartículas/química , Paclitaxel/uso terapéutico , Polietilenglicoles/química , Profármacos/farmacología , Profármacos/uso terapéutico , Distribución Tisular
11.
Mol Pharm ; 16(8): 3489-3501, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31246475

RESUMEN

Recently, multifunctional clearable inorganic theranostic nanoparticles have been attracting more and more attention. Protein-based nanoparticles can be cleared by the hepatobiliary system efficiently. In this work, ultrasmall gadolinium oxide (Gd2O3) nanoparticles, which possess the advantage of high longitudinal relaxation rate, were coated with bovine serum albumin (BSA). After the Gd2O3/BSA nanoparticles were linked with two-dimensional photothermal MoS2 nanomaterials, the nanoparticles were also modified with hyaluronic acid (HA) through the disulfide bonds for tumor-targeting effect. As indicated by in vitro and in vivo studies, these Gd2O3/BSA@MoS2-HA nanoparticles could be rapidly degraded and excreted after reacting with glutathione (GSH) by the redox response, thus avoiding long-term toxicity. In addition, the cellular uptake study and in vivo multispectral optoacoustic tomography (MSOT), X-ray computed tomography (CT), and magnetic resonance (MR) triple-modal images demonstrated that Gd2O3/BSA@MoS2-HA nanoparticles exhibited a high tumor uptake effect after intravenous injection. Consequently, such clearable theranostic nanoparticles with multiple functions, which are applicable in multimodal imaging-guided cancer therapy, might show promise for applications in nanomedical science.


Asunto(s)
Gadolinio/administración & dosificación , Imagen Multimodal/métodos , Nanopartículas/administración & dosificación , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animales , Línea Celular Tumoral/trasplante , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Gadolinio/química , Gadolinio/farmacocinética , Glutatión/metabolismo , Humanos , Hipertermia Inducida/métodos , Inyecciones Intravenosas , Imagen por Resonancia Magnética/métodos , Tasa de Depuración Metabólica , Ratones , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Tamaño de la Partícula , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Albúmina Sérica Bovina/química , Distribución Tisular , Tomografía Computarizada por Rayos X/métodos
12.
J Pharm Sci ; 108(9): 3124-3129, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31136767

RESUMEN

Microdialysis is a technique that utilizes a semipermeable membrane to sample analytes present within tissue interstitial fluid. Analyte-specific calibration is required for quantitative microdialysis, but these calibration methods are tedious, require significant technical skill, and often cannot be performed jointly with the experimental measurements. Here, we describe a method using retrodialysis with stable-isotope-labeled analytes that enables simultaneous calibration and quantification for in vivo tumor microdialysis. Isotope-labeled amino acids relevant to immuno-metabolism in the tumor microenvironment (tryptophan, kynurenine, glutamine, and glutamate) were added to the microdialysis perfusate, and microdialysis probes were inserted in subcutaneous CT26 and MC38 tumors in mice. The levels of both the endogenous and isotope-labeled amino acids in the perfusate outlet were quantified using LC-MS/MS. Plasma and tumor tissue samples were also collected from the same mice and amino acid levels quantified using LC-MS/MS. Amino acids which showed statistically significant differences between the CT26-bearing and MC38-bearing mice in tumor lysate (tryptophan, kynurenine, and glutamine) and plasma (glutamate) were not the same as those identified as significantly different in tumor interstitial fluid (kynurenine and glutamate), underscoring how microdialysis can provide unique and complementary insights into tumor and immune metabolism within the tumor microenvironment.


Asunto(s)
Aminoácidos/análisis , Antineoplásicos/farmacología , Monitoreo de Drogas/métodos , Microdiálisis/normas , Microambiente Tumoral/efectos de los fármacos , Aminoácidos/metabolismo , Animales , Antineoplásicos/uso terapéutico , Calibración , Línea Celular Tumoral/trasplante , Cromatografía Líquida de Alta Presión/métodos , Soluciones para Diálisis/análisis , Soluciones para Diálisis/química , Modelos Animales de Enfermedad , Femenino , Humanos , Marcaje Isotópico , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Microdiálisis/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Espectrometría de Masas en Tándem/métodos
13.
Sci Rep ; 9(1): 4712, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886310

RESUMEN

The recent past has seen impressive progress in the treatment of various malignancies using immunotherapy. One of the most promising approaches involves immune checkpoint inhibitors. However, the clinical results with these agents have demonstrated variability in the response. Pancreatic cancer, in particular, has proven resistant to initial immunotherapy approaches. Here, we describe an alternative strategy that relies on combining gemcitabine and a novel programmed death-ligand 1 (PD-L1) inhibitor, termed MN-siPDL1. MN-siPDL1 incorporates small interfering RNA against PD-L1 (siPDL1) conjugated to a magnetic nanocarrier (MN). We show that noninvasive magnetic resonance imaging (MRI) could be used to monitor therapeutic response. Combination therapy consisting of gemcitabine and MN-siPDL1 in a syngeneic murine pancreatic cancer model resulted in a significant reduction in tumor growth and an increase in survival. Following optimization, a 90% reduction in tumor volume was achieved 2 weeks after the beginning of treatment. Whereas 100% of the control animals had succumbed to their tumors by week 6 after the beginning of treatment, there was no mortality in the experimental group by week 5, and 67% of the experimental animals survived for 12 weeks. This method could provide therapeutic benefit against an intractable disease for which there are no effective treatments and which is characterized by a mere 1% 5-year survival.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma Ductal Pancreático/tratamiento farmacológico , Portadores de Fármacos/química , Inmunoterapia/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/farmacología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Carcinoma Ductal Pancreático/diagnóstico por imagen , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral/trasplante , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Monitoreo de Drogas/métodos , Femenino , Humanos , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/química , Dosis Máxima Tolerada , Ratones , Páncreas/diagnóstico por imagen , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Interferencia de ARN , ARN Interferente Pequeño/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Gemcitabina
14.
Nat Commun ; 10(1): 1192, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30867429

RESUMEN

Development of photothermal materials which are able to harness sunlight and convert it to thermal energy seems attractive. Besides carbon-based nanomaterials, conjugated polymers are emerging promising photothermal materials but their facile syntheses remain challenging. In this work, by modification of a CBT-Cys click condensation reaction and rational design of the starting materials, we facilely synthesize conjugated polymers poly-2-phenyl-benzobisthiazole (PPBBT) and its dihexyl derivative with good photothermal properties. Under the irradiation of either sunlight-mimicking Xe light or near-infrared laser, we verify that PPBBT has comparable photothermal heating-up speed to that of star material single-wall carbon nanotube. Moreover, PPBBT is used to fabricate water-soluble NPPPBBT nanoparticles which maintain excellent photothermal properties in vitro and photothermal therapy effect on the tumours exposed to laser irradiation. We envision that our synthetic method provides a facile approach to fabricate conjugated polymers for more promising applications in biomedicine or photovoltaics in the near future.


Asunto(s)
Hipertermia Inducida/métodos , Nanopartículas/efectos de la radiación , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Terapia Ultravioleta/métodos , Animales , Línea Celular Tumoral/trasplante , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertermia Inducida/instrumentación , Rayos Láser , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Nanopartículas/administración & dosificación , Nanopartículas/química , Nanopartículas/ultraestructura , Polímeros/administración & dosificación , Polímeros/química , Polímeros/farmacocinética , Polímeros/efectos de la radiación , Distribución Tisular , Resultado del Tratamiento , Terapia Ultravioleta/instrumentación
15.
J Surg Res ; 233: 310-322, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30502264

RESUMEN

BACKGROUND: Reproduction of the perfusion used in therapy (hyperthermic intraperitoneal chemotherapy) procedures preclinically represents a valuable asset for investigating new therapeutic agents that may improve patient outcomes. This article provides technical descriptions of our execution of closed and open "coliseum" abdominal perfusion techniques in a mouse model of peritoneal carcinomatosis of colorectal cancer. MATERIALS AND METHODS: BALB/c mice presenting with disseminated colorectal cancer (CT26-luciferin cells) underwent 30-min perfusions mimicking either the closed perfusion or the coliseum perfusion technique. Disease burden was monitored by bioluminescence signaling using an in vivo imaging system. Perfusion circuits consisted of single inflow lines with either a single or dual outflow line. RESULTS: Twelve mice presenting with disseminated disease underwent the closed perfusion technique. Surgical complications included perfusate leakage and organ constriction/suction into the outflow line(s). Nine mice underwent the coliseum perfusion technique with surgical debulking, using bipolar cauterization to remove tumors attached to the peritoneum. All mice survived the coliseum perfusion with limited intraoperative complications. CONCLUSIONS: Fewer intraoperative complications were experienced with our coliseum perfusion technique than the closed perfusion. The methods described here can be used as a guideline for developing future perfusion murine models for investigating perfusion models useful for delivery of chemotherapy or other tumor-sensitization agents, including selective targeted agents, nanoparticles, and heat.


Asunto(s)
Antineoplásicos/administración & dosificación , Quimioterapia del Cáncer por Perfusión Regional/métodos , Neoplasias Colorrectales/terapia , Hipertermia Inducida/métodos , Neoplasias Peritoneales/terapia , Animales , Línea Celular Tumoral/trasplante , Quimioterapia del Cáncer por Perfusión Regional/efectos adversos , Quimioterapia del Cáncer por Perfusión Regional/instrumentación , Neoplasias Colorrectales/patología , Terapia Combinada/efectos adversos , Terapia Combinada/instrumentación , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertermia Inducida/efectos adversos , Hipertermia Inducida/instrumentación , Complicaciones Intraoperatorias/epidemiología , Complicaciones Intraoperatorias/etiología , Ratones , Ratones Endogámicos BALB C , Neoplasias Peritoneales/secundario , Resultado del Tratamiento
16.
Mol Pharm ; 15(7): 2594-2605, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29763568

RESUMEN

We previously developed a new zinc(II) phthalocyanine (ZnPc) derivative (Pc 1) conjugated to poly-L-glutamic acid (PGA) (1-PG) to address the limitations of ZnPc as part of an antitumor photodynamic therapy approach, which include hydrophobicity, phototoxicity, and nonselectivity in biodistribution and tumor targeting. During this study, we discovered that 1-PG possessed high near-infrared (NIR) light absorptivity (λmax = 675 nm), good singlet oxygen generation efficiency in an aqueous environment, and enhanced photocytotoxic efficacy and cancer cell uptake in vitro. In the current study, we discovered that 1-PG accumulated in 4T1 mouse mammary tumors, with a retention time of up to 48 h. Furthermore, as part of an antitumor PDT, low dose 1-PG (2 mg of Pc 1 equivalent/kg) induced a greater tumor volume reduction (-74 ± 5%) when compared to high dose ZnPc (8 mg/kg, -50 ± 12%). At higher treatment doses (8 mg of Pc 1 equivalent/kg), 1-PG reduced tumor volume maximally (-91 ± 6%) and suppressed tumor size to a minimal level for up to 15 days. The kidney, liver, and lungs of the mice treated with 1-PG (both low and high doses) were free from 4T1 tumor metastasis at the end of the study. Telemetry-spectral-echocardiography studies also revealed that PGA (65 mg/kg) produced insignificant changes to the cardiovascular physiology of Wistar-Kyoto rats when administered in vivo. Results indicate that PGA displays an excellent cardiovascular safety profile, underlining its suitability for application as a nanodrug carrier in vivo. These current findings indicate the potential of 1-PG as a useful photosensitizer candidate for clinical PDT.


Asunto(s)
Indoles/administración & dosificación , Nanoconjugados/química , Neoplasias/tratamiento farmacológico , Compuestos Organometálicos/administración & dosificación , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Cardiotoxicidad/diagnóstico por imagen , Cardiotoxicidad/etiología , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Ecocardiografía , Humanos , Indoles/efectos adversos , Indoles/farmacocinética , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/patología , Compuestos Organometálicos/efectos adversos , Compuestos Organometálicos/farmacocinética , Fotoquimioterapia/efectos adversos , Fármacos Fotosensibilizantes/efectos adversos , Fármacos Fotosensibilizantes/farmacocinética , Ácido Poliglutámico/química , Ratas , Ratas Endogámicas WKY , Distribución Tisular
17.
Cancer Res ; 78(13): 3698-3708, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29735553

RESUMEN

Although malignant phenotypes of triple-negative breast cancer (TNBC) are subject to circadian alterations, the role of cancer stem cells (CSC) in defining this circadian change remains unclear. CSC are often characterized by high aldehyde dehydrogenase (ALDH) activity, which is associated with the malignancy of cancer cells and is used for identification and isolation of CSC. Here, we show that the population of ALDH-positive cells in a mouse 4T1 breast tumor model exhibits pronounced circadian alterations. Alterations in the number of ALDH-positive cells were generated by time-dependent increases and decreases in the expression of Aldh3a1 Importantly, circadian clock genes were rhythmically expressed in ALDH-negative cells, but not in ALDH-positive cells. Circadian expression of Aldh3a1 in ALDH-positive cells was dependent on the time-dependent release of Wingless-type mmtv integration site family 10a (WNT10a) from ALDH-negative cells. Furthermore, antitumor and antimetastatic effects of ALDH inhibitor N,N-diethylaminobenzaldehyde were enhanced by administration at the time of day when ALDH activity was increased in 4T1 tumor cells. Our findings reveal a new role for the circadian clock within the tumor microenvironment in regulating the circadian dynamics of CSC. These results should enable the development of novel therapeutic strategies for treatment of TNBC with ALDH inhibitors.Significance: This seminal report reveals that circadian dynamics of CSC are regulated by the tumor microenvironment and provides a proof of principle of its implication for chronotherapy in TNBC. Cancer Res; 78(13); 3698-708. ©2018 AACR.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Benzaldehídos/administración & dosificación , Relojes Circadianos/fisiología , Células Madre Neoplásicas/fisiología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Aldehído Deshidrogenasa/metabolismo , Animales , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Proteínas Wnt/metabolismo
18.
Cancer Lett ; 428: 21-33, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29702192

RESUMEN

Ferroptosis is a form of programmed cell death decided by iron-dependent lipid peroxidation, but its role in glioma cell death remains unclear. In this study, we found Pseudolaric acid B (PAB) inhibited the viabilities of glioma cells in vitro and in vivo, which was accompanied by abnormal increases of intracellular ferrous iron, H2O2 and lipid peroxidation, as well as depletion of GSH and cysteine. In vitro studies revealed that the lipid peroxidation and the cell death caused by PAB were both inhibited by iron chelator deferoxamine, but exacerbated by supplement of ferric ammonium citrate. Inhibition of lipid peroxidation with ferrostatin-1 or GSH rescued PAB-induced cell death. Morphologically, the cells treated with PAB presented intact membrane, shrunken mitochondria with increased membrane density, and normal-sized nucleus without chromatin condensation. Mechanistically, PAB improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2 and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT pathway, which further exacerbated accumulation of H2O2 and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Diterpenos/farmacología , Glioma/tratamiento farmacológico , Peroxidación de Lípido/efectos de los fármacos , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral/trasplante , Ciclohexilaminas/farmacología , Modelos Animales de Enfermedad , Diterpenos/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADPH Oxidasa 4/metabolismo , Fenilendiaminas/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Cancer Prev Res (Phila) ; 11(7): 383-392, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29636351

RESUMEN

Three decades ago, the Garlands postulated that vitamin D3 produced in the skin by ultraviolet radiation (UVR)-induced conversion of 7-dehydrocholesterol to pre-D3 has anticancer effects, thus triggering more than 9,500 publications on D3 and cancer. Here, we report that UVR treatment of transgenic mice of the well-established C3(1)/SV40 Tag mammary cancer model significantly inhibits both autochthonous carcinogenesis and allograft tumor growth, but in contrast neither dietary nor topical D3 influences mammary carcinogenesis in this specific mouse model. Furthermore, UVR's inhibitory effects occur irrespective of whether or not the treatment increases circulating D3 in the mice. The inhibitory effect of UVR on autochthonous tumors occurs at or before the stage of ductal carcinoma in situ. Our studies indicate clearly that UVR can exert D3-independent anticancer effects in C3(1)/SV40 Tag mice. Therefore, supplemental D3 may not mimic all possible beneficial effects of UVR, and uncovering non-D3-mediated mechanisms of UVR tumor inhibition may lead to novel strategies for cancer prevention. Cancer Prev Res; 11(7); 383-92. ©2018 AACR.


Asunto(s)
Carcinogénesis/efectos de la radiación , Carcinoma Intraductal no Infiltrante/prevención & control , Neoplasias Mamarias Experimentales/prevención & control , Receptores de Estrógenos/metabolismo , Rayos Ultravioleta , Animales , Carcinoma Intraductal no Infiltrante/patología , Línea Celular Tumoral/trasplante , Colecalciferol/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Piel/metabolismo , Piel/efectos de la radiación
20.
Pharm Res ; 35(4): 80, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500548

RESUMEN

PURPOSE: Synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs were found to be able to target cells that express Toll-like receptor 9 to modulate innate and adaptive immune reactions. But their in vivo application in immunotherapy against cancer has not been successful. We attempted in this study to examine polyethylene-glycol (PEG) conjugated CpG ODNs and investigated their mechanism of immune modulation in anti-cancer therapy. METHODS: CpG-PEG conjugates with different PEG lengths were synthesized. In vitro activity as well as in vivo pharmacokinetics and pharmacodynamics properties were evaluated. RESULTS: CpG-PEG20Ks were found to be able to persist longer in circulation and activate various downstream effector cells. After intravenous injection, they resulted in higher levels of IL-12p70 in the circulation and lower M-MDSC infiltrates in the tumor microenvironment. Such activities were different from those of CpG ODNs without PEGylation, suggesting different PK-PD profiles systemically and locally. CONCLUSIONS: Our data support the development of CpG-PEGs as a new therapeutic agent that can be systemically administered to modulate immune responses and the microenvironment in tumor tissues.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Composición de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Oligodesoxirribonucleótidos/farmacología , Animales , Línea Celular Tumoral/trasplante , Células Cultivadas , Células Dendríticas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inyecciones Intravenosas , Interleucina-12/inmunología , Interleucina-12/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/uso terapéutico , Ovalbúmina/inmunología , Polietilenglicoles/química , Cultivo Primario de Células , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA