Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.011
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Cardiovasc Res ; 118(18): 3499-3516, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36461918

RESUMEN

For many patients with terminal/advanced cardiac failure, heart transplantation is the most effective, durable treatment option, and offers the best prospects for a high quality of life. The number of potentially life-saving donated human organs is far fewer than the population who could benefit from a new heart, resulting in increasing numbers of patients awaiting replacement of their failing heart, high waitlist mortality, and frequent reliance on interim mechanical support for many of those deemed among the best candidates but who are deteriorating as they wait. Currently, mechanical assist devices supporting left ventricular or biventricular heart function are the only alternative to heart transplant that is in clinical use. Unfortunately, the complication rate with mechanical assistance remains high despite advances in device design and patient selection and management, and the quality of life of the patients even with good outcomes is only moderately improved. Cardiac xenotransplantation from genetically multi-modified (GM) organ-source pigs is an emerging new option as demonstrated by the consistent long-term success of heterotopic (non-life-supporting) abdominal and life-supporting orthotopic porcine heart transplantation in baboons, and by a recent 'compassionate use' transplant of the heart from a GM pig with 10 modifications into a terminally ill patient who survived for 2 months. In this review, we discuss pig heart xenotransplantation as a concept, including pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental overgrowth of the heart, as well as GM strategies in pigs to prevent or minimize these problems. Additional topics discussed include relevant results of heterotopic and orthotopic heart transplantation experiments in the pig-to-baboon model, microbiological and virologic safety concepts, and efficacy requirements for initiating formal clinical trials. An adequate regulatory and ethical framework as well as stringent criteria for the selection of patients will be critical for the safe clinical development of cardiac xenotransplantation, which we expect will be clinically tested during the next few years.


Asunto(s)
Trasplante de Corazón , Calidad de Vida , Humanos , Animales , Porcinos , Trasplante Heterólogo/efectos adversos , Trasplante Heterólogo/métodos , Trasplante de Corazón/efectos adversos , Resultado del Tratamiento , Rechazo de Injerto/prevención & control , Animales Modificados Genéticamente
2.
Cells ; 13(1)2023 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-38201286

RESUMEN

Hepatocellular carcinoma (HCC) results in high mortality due to ineffective systemic therapy. Human immortalized cell lines are commonly used to study anti-tumor effects in the context of new anti-tumor therapies and tumor biology. As immortalized cell lines have limited biological relevance and heterogeneity compared to primary cells, patient-derived tumor tissues, and corresponding immune cells are the gold standards for studying the complexity of individual tumor entities. However, culturing primary HCC cells has a low success rate. Here, we aimed to establish a reproducible approach to preserve the patient-derived liver cancer cells for in vitro and in vivo studies. The underlying study aimed to establish an in vitro pre-screening platform to test treatment options' effectivity and dosage, e.g., for new substances, autologous modified immune cells, or combined therapies in HCC. We initially employed 15 surgical resection specimens from patients with different HCC entities for isolation and preservation. The isolated liver cancer cells from four HCC-diagnosed patients were used for orthotopic transplantation into the healthy liver of immunodeficient mice, allowing them to grow for six months before human liver cancer cells were isolated and cultured. As a result, we generated and characterized four new primary-like liver cancer cell lines. Compared to immortalized HCC cell lines, freshly generated liver cancer cells displayed individual morphologies and heterogeneous protein-level characteristics. We assessed their ability to proliferate, migrate, form spheroids, and react to common medications compared to immortalized HCC cell lines. All four liver cancer cell lines exhibit strong migration and colony-forming characteristics in vitro, comparable to extensively investigated immortalized HCC cell lines. Moreover, the four etiological different liver cancer cell lines displayed differences in the response to 5-FU, Sorafenib, Axitinib, and interferon-alpha treatment, ranking from non-responders to responders depending on the applicated medication. In sum, we generated individual patient-derived liver cancer cell lines suitable for predictive in vitro drug screenings and for xenograft transplantations to realize the in vivo investigation of drug candidates. We overcame the low cultivation success rate of liver cancer cells derived from patients and analyzed their potential to serve a pre-clinical model.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animales , Ratones , Carcinoma Hepatocelular/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Neoplasias Hepáticas/tratamiento farmacológico , Trasplante Heterólogo , Línea Celular
3.
Diabetes Metab J ; 46(5): 677-688, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35124687

RESUMEN

BACKGROUND: Neonatal porcine pancreatic cell clusters (NPCCs) have been proposed as an alternative source of ß cells for islet transplantation because of their low cost and growth potential after transplantation. However, the delayed glucose lowering effect due to the immaturity of NPCCs and immunologic rejection remain as a barrier to NPCC's clinical application. Here, we demonstrate accelerated differentiation and immune-tolerant NPCCs by in vitro chemical treatment and microencapsulation. METHODS: NPCCs isolated from 3-day-old piglets were cultured in F-10 media and then microencapsulated with alginate on day 5. Differentiation of NPCCs is facilitated by media supplemented with activin receptor-like kinase 5 inhibitor II, triiodothyronine and exendin-4 for 2 weeks. Marginal number of microencapsulated NPCCs to cure diabetes with and without differentiation were transplanted into diabetic mice and observed for 8 weeks. RESULTS: The proportion of insulin-positive cells and insulin mRNA levels of NPCCs were significantly increased in vitro in the differentiated group compared with the undifferentiated group. Blood glucose levels decreased eventually after transplantation of microencapsulated NPCCs in diabetic mice and normalized after 7 weeks in the differentiated group. In addition, the differentiated group showed nearly normal glucose tolerance at 8 weeks after transplantation. In contrast, neither blood glucose levels nor glucose tolerance were improved in the undifferentiated group. Retrieved graft in the differentiated group showed greater insulin response to high glucose compared with the undifferentiated group. CONCLUSION: in vitro differentiation of microencapsulated immature NPCCs increased the proportion of insulin-positive cells and improved transplant efficacy in diabetic mice without immune rejection.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Alginatos/metabolismo , Alginatos/farmacología , Animales , Animales Recién Nacidos , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/cirugía , Exenatida/farmacología , Insulina/metabolismo , Ratones , ARN Mensajero/metabolismo , ARN Mensajero/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Porcinos , Trasplante Heterólogo , Triyodotironina/metabolismo , Triyodotironina/farmacología
4.
J Crohns Colitis ; 16(1): 109-121, 2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-34180971

RESUMEN

BACKGROUND AND AIMS: Ulcerative colitis [UC] is a chronic inflammatory disease of the colon with frequent relapses. Telomere shortening in intestinal epithelial cells has been reported in severe or longstanding cases. However, its influence on UC pathogenesis remains unelucidated. To this end, we evaluated telomere shortening using a long-term organoid inflammation model that we had originally established. METHODS: A UC model using human colon organoids was established to assess telomere changes chronologically. MST-312 was used for the telomerase inhibition assay. The potential of telomerase activators as a novel UC treatment was evaluated with an in vitro model, including microarray analysis, and histological changes were assessed using xenotransplantation into mouse colonic mucosa. RESULTS: Our UC model reproduced telomere shortening in vitro, which was induced by the continuous suppression of telomerase activity via P53. MST-312-based analysis revealed that telomere shortening was involved in the pathogenesis of UC. Madecassoside [MD] improved the telomere length of the UC model and UC patient-derived organoids, which further promoted cell proliferation in vitro and improved the graft take-rate of xenotransplantation. Moreover, histological analysis revealed that MD induced normal crypt structure with abundant goblet cells. CONCLUSIONS: This study is the first to reveal the mechanism and importance of telomere shortening in the pathogenesis of UC. MD could be a novel candidate for UC treatment beyond endoscopic mucosal healing.


Asunto(s)
Colitis Ulcerosa/patología , Células Epiteliales/patología , Mucosa Intestinal/citología , Acortamiento del Telómero , Animales , Biopsia , Proliferación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colonoscopía , Humanos , Ratones , Organoides/metabolismo , Organoides/patología , Organoides/trasplante , Especies Reactivas de Oxígeno/metabolismo , Telomerasa/metabolismo , Trasplante Heterólogo
5.
J Nanobiotechnology ; 19(1): 433, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930269

RESUMEN

BACKGROUND: The construction of a nanoimmune controlled-release system that spatiotemporally recognizes tumor lesions and stimulates the immune system response step by step is one of the most potent cancer treatment strategies for improving the sensitivity of immunotherapy response. RESULTS: Here, a composite nanostimulator (CNS) was constructed for the release of second near-infrared (NIR-II) photothermal-mediated immune agents, thereby achieving spatiotemporally controllable photothermal-synergized immunotherapy. CNS nanoparticles comprise thermosensitive liposomes as an outer shell and are internally loaded with a NIR-II photothermal agent, copper sulfide (CuS), toll-like receptor-9 (TLR-9) agonist, cytosine-phospho-guanine oligodeoxynucleotides, and programmed death-ligand 1 (PD-L1) inhibitors (JQ1). Following NIR-II photoirradiation, CuS enabled the rapid elevation of localized temperature, achieving tumor ablation and induction of immunogenic cell death (ICD) as well as disruption of the lipid shell, enabling the precise release of two immune-therapeutical drugs in the tumor region. Combining ICD, TLR-9 stimulation, and inhibited expression of PD-L1 allows the subsequent enhancement of dendritic cell maturation and increases infiltration of cytotoxic T lymphocytes, facilitating regional antitumor immune responses. CONCLUSION: CNS nanoparticle-mediated photothermal-synergized immunotherapy efficiently suppressed the growth of primary and distant tumors in two mouse models and prevented pulmonary metastasis. This study thus provides a novel sight into photo-controllably safe and efficient immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Rayos Infrarrojos , Nanopartículas/química , Neoplasias/terapia , Fototerapia/métodos , Animales , Azepinas/química , Azepinas/farmacología , Azepinas/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Cobre/química , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Verde de Indocianina/química , Verde de Indocianina/uso terapéutico , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor Toll-Like 9/metabolismo , Trasplante Heterólogo , Triazoles/química , Triazoles/farmacología , Triazoles/uso terapéutico
6.
J Nanobiotechnology ; 19(1): 432, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930301

RESUMEN

BACKGROUND: Distant metastasis to vital organs is the major contributor to breast cancer mortality, and regional lymph node metastasis is an important facilitator of distant metastasis and recurrence in this cancer. The early diagnosis and precise treatment of lymph node metastasis are crucial for staging and prognosis in breast cancer. Herein, we report a visualized precision medicine nanoplatform of metastatic lymph nodes for ultrasonic/photoacoustic (US/PA) dual modal imaging-guided in situ targeted hyperthermia-combined chemotherapy. RESULTS: Carbon nanoparticles (CNs), approved by the China Food and Drug Administration, were loaded with docetaxel and rationally combined with anti-hypoxia-inducible factor 1α antibody-modified poly (lactic-co-glycolic acid) (PLGA) nanoparticles to achieve the combination of passive targeting at the lymph nodes and intracellular targeting at HIF 1α factor. The accumulation and retention of nanoparticles in metastatic lymph nodes via lymphatic delivery were enhanced. Docetaxel could be effectively offloaded by CNs that have active carbon nanoparticles, and the PLGA membrane prevented drug leakage. The nanoparticles exhibited excellent photothermal performance with a photothermal conversion efficiency of 28.9%, killing tumor cells in metastatic lymph nodes through hyperthermia. In vitro and in vivo systematic evaluations revealed that hyperpyrexia triggered the rupture of nanoparticles caused by the phase transition of perfluorohexane, resulting in docetaxel release for achieving in situ hyperthermia-combined chemotherapy. CONCLUSIONS: The laser-triggered highly efficient in situ chemotherapy nanosystem achieves targeted synergistic chemo-hyperthermia treatment of metastatic lymph nodes, and lymphatic delivery represents a strategy to avoid additional injury caused by drugs entering the blood circulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Hipertermia Inducida/métodos , Ganglios Linfáticos/metabolismo , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos/química , Anticuerpos/inmunología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carbono/química , Línea Celular Tumoral , Docetaxel/química , Docetaxel/metabolismo , Docetaxel/farmacología , Docetaxel/uso terapéutico , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Rayos Infrarrojos , Metástasis Linfática , Nanomedicina , Nanopartículas/metabolismo , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratas , Trasplante Heterólogo
7.
Chem Commun (Camb) ; 57(59): 7296-7299, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34223569

RESUMEN

A chlorine e6 (Ce6) and curcumin (Cur) based self-delivery nanomedicine (CeCu) is prepared for chemotherapy sensitized photodynamic therapy (PDT). The chemotherapeutic agent of Cur could inhibit the TrxR activity to destroy the cellular ROS-defence system for enhanced PDT, which provides synergistic effects for tumor precision therapy in consideration of the unfavorable tumor microenvironments.


Asunto(s)
Nanomedicina , Fotoquimioterapia/métodos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Clorofilidas , Curcumina/química , Curcumina/metabolismo , Curcumina/farmacología , Humanos , Ratones , Microscopía Confocal , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/química , Porfirinas/metabolismo , Porfirinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Trasplante Heterólogo , Microambiente Tumoral
8.
Adv Mater ; 33(37): e2102054, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34309925

RESUMEN

Arsenical drugs have achieved hallmark success in treating patients with acute promyelocytic leukemia, but expanding their clinical utility to solid tumors has proven difficult with the contradiction between the therapeutic efficacy and the systemic toxicity. Here, leveraging efforts from materials science, biocompatible PEGylated arsenene nanodots (AsNDs@PEG) with high monoelemental arsenic purity that can selectively and effectively treat solid tumors are synthesized. The intrinsic selective killing effect of AsNDs@PEG is closely related to high oxidative stress in tumor cells, which leads to an activated valence-change of arsenic (from less toxic As0 to severely toxic oxidation states), followed by decreased superoxide dismutase activity and massive reactive oxygen species (ROS) production. These effects occur selectively within cancer cells, causing mitochondrial damage, cell-cycle arrest, and DNA damage. Moreover, AsNDs@PEG when applied in a multi-drug combination strategy with ß-elemene, a plant-derived anticancer drug, achieves synergistic antitumor outcomes, and its newly discovered on-demand photothermal properties facilitate the elimination of the tumors without recurrence, potentially further expanding its clinical utility. In line of the practicability for a large-scale fabrication and negligible systemic toxicity of AsNDs@PEG (even at high doses and with repetitive administration), a new-concept arsenical drug with high therapeutic efficacy for selective solid tumor therapy is provided.


Asunto(s)
Antineoplásicos/farmacología , Arsénico/química , Nanopartículas/química , Sesquiterpenos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Quimioterapia Combinada , Humanos , Rayos Infrarrojos , Ratones , Ratones Desnudos , Nanopartículas/uso terapéutico , Nanopartículas/toxicidad , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , Terapia Fototérmica , Polietilenglicoles/química , Especies Reactivas de Oxígeno/metabolismo , Sesquiterpenos/química , Sesquiterpenos/uso terapéutico , Trasplante Heterólogo
9.
J Photochem Photobiol B ; 221: 112257, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34271410

RESUMEN

Organic semiconductor small molecules IHIC and ITIC have been developed as solar cell materials, and because of their strong near-infrared absorption capabilities, they are promising for cancer phototherapy. This article reports the application of semiconductor small molecule IHIC/ITIC liposomes in photothermal therapy and photoacoustic imaging of tumors firstly. Experiments show that the liposome-loaded IHIC/ITIC material has good biocompatibility and can be effectively enriched in tumor sites. After being irradiated with laser, it can emit strong photoacoustic signals, and has achieved good results in the photothermal treatment of breast cancer mice. We believe that organic semiconductor small molecule IHIC/ITIC will become a promising photothermal agent with wonderful development possibilities.


Asunto(s)
Materiales Biocompatibles/química , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Semiconductores , Animales , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/efectos de la radiación , Células HeLa , Humanos , Rayos Láser , Liposomas/química , Ratones , Ratones Desnudos , Microscopía Confocal , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Tamaño de la Partícula , Técnicas Fotoacústicas , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/farmacología , Terapia Fototérmica , Trasplante Heterólogo
10.
Chem Commun (Camb) ; 57(59): 7240-7243, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34190264

RESUMEN

Herin, we report a Cu(ii)-porphyrin-derived nanoscale COF, which can be triggered by endogenous H2S via an intracellular sulfidation reaction to generate a metal-free COF-photosensitizer for PDT against H2S-enriched colon tumors with controllable singlet oxygen release; meanwhile in situ generated CuS can be synchronously used as a photothermal agent for PTT.


Asunto(s)
Neoplasias del Colon/terapia , Sulfuro de Hidrógeno/química , Estructuras Metalorgánicas/química , Fototerapia/métodos , Animales , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Cobre/química , Células HCT116 , Humanos , Rayos Infrarrojos , Ratones , Microscopía Confocal , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/química , Oxígeno Singlete/metabolismo , Trasplante Heterólogo
11.
Reprod Sci ; 28(11): 3109-3122, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34008154

RESUMEN

This study evaluated the effect of adding alpha lipoic acid (ALA) to the vitrification solution of sheep ovarian tissue on 7 days of in vitro culture or 15 days of xenotransplantion. ALA was used at two different concentrations (100 µM: ALA100 and 150 µM: ALA150). Ovarian tissue was evaluated by classical histology (follicular morphology, development, and stromal cell density); immunohistochemistry for forkhead box O3a (FOXO3a); Ki67 (cell proliferation); cluster of differentiation 31 (CD31); and alpha smooth muscle actin (α-SMA). Reactive oxygen species (ROS) levels in ovarian tissue, as well as malondialdehyde (MDA) and nitrite levels in the culture medium, were assessed. Similar percentage of morphologically normal follicles was found in the vitrified ovarian tissue in the presence of ALA100 or ALA150 after in vitro culture or xenotransplantation. Follicular development from all treatments was higher (P < 0.05) than the control group. Moreover, an activation of primordial follicles was observed by FOXO3a. Stromal cell density and immunostaining for Ki67 and CD31 were significantly higher (P < 0.05) in ALA150 vitrified tissue. No difference (P > 0.05) was found in α-SMA between ALA concentrations after in vitro culture or xenograft. ROS levels in the ovarian tissue were similar (P > 0.05) in all treatments, as well as MDA and nitrite levels after 7 days of culture. We concluded that the addition of ALA 150 is able to better preserve the stromal cell density favoring granulosa cell proliferation and neovascularization.


Asunto(s)
Antioxidantes/farmacología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/trasplante , Ácido Tióctico/farmacología , Trasplante Heterólogo/métodos , Vitrificación/efectos de los fármacos , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Folículo Ovárico/fisiología , Ovario/efectos de los fármacos , Ovario/fisiología , Ovario/trasplante , Ovinos
12.
J Med Chem ; 64(10): 6877-6901, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33999621

RESUMEN

BRAF is an important component of MAPK cascade. Mutation of BRAF, in particular V600E, leads to hyperactivation of the MAPK pathway and uncontrolled cellular growth. Resistance to selective inhibitors of mutated BRAF is a major obstacle against treatment of many cancer types. In this work, a series of new (imidazo[2,1-b]thiazol-5-yl)pyrimidine derivatives possessing a terminal sulfonamide moiety were synthesized. Pan-RAF inhibitory effect of the new series was investigated, and structure-activity relationship is discussed. Antiproliferative activity of the target compounds was tested against the NCI-60 cell line panel. The most active compounds were further tested to obtain their IC50 values against cancer cells. Compound 27c with terminal open chain sulfonamide and 38a with a cyclic sulfamide moiety showed the highest activity in enzymatic and cellular assay, and both compounds were able to inhibit phosphorylation of MEK and ERK. Compound 38a was selected for testing its in vivo activity against melanoma. Cellular and animal activities are reported.


Asunto(s)
Imidazoles/química , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Tiazoles/química , Animales , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Semivida , Humanos , Imidazoles/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/metabolismo , Relación Estructura-Actividad , Sulfonamidas/química , Tiazoles/metabolismo , Trasplante Heterólogo
13.
Angew Chem Int Ed Engl ; 60(24): 13405-13413, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33755286

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by an aberrant metabolic phenotype with high metastatic capacity, resulting in poor patient prognoses and low survival rates. We designed a series of novel AuIII cyclometalated prodrugs of energy-disrupting Type II antidiabetic drugs namely, metformin and phenformin. Prodrug activation and release of the metformin ligand was achieved by tuning the cyclometalated AuIII fragment. The lead complex 3met was 6000-fold more cytotoxic compared to uncoordinated metformin and significantly reduced tumor burden in mice with aggressive breast cancers with lymphocytic infiltration into tumor tissues. These effects was ascribed to 3met interfering with energy production in TNBCs and inhibiting associated pro-survival responses to induce deadly metabolic catastrophe.


Asunto(s)
Antineoplásicos/metabolismo , Metformina/metabolismo , Profármacos/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/química , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Oro/química , Humanos , Metformina/química , Ratones , Conformación Molecular , Fenformina/química , Fenformina/metabolismo , Profármacos/química , Profármacos/farmacología , Profármacos/uso terapéutico , Trasplante Heterólogo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
14.
Eur J Med Chem ; 216: 113307, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33652354

RESUMEN

Androgen receptor (AR) is an effective therapeutic target for the treatment of prostate cancer. We report herein the design, synthesis, and biological evaluation of highly effective proteolysis targeting chimeras (PROTAC) androgen receptor (AR) degraders, such as compound A031. It could induce the degradation of AR protein in VCaP cell lines in a time-dependent manner, achieving the IC 50 value of less than 0.25 µM. The A031 is 5 times less toxic than EZLA and works with an appropriate half-life (t 1/2) or clearance rate (Cl). Also, it has a significant inhibitory effect on tumor growth in zebrafish transplanted with human prostate cancer (VCaP). Therefore, A031 provides a further idea of developing novel drugs for prostate cancer.


Asunto(s)
Antagonistas de Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/farmacocinética , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/genética , Relación Estructura-Actividad , Tasa de Supervivencia , Trasplante Heterólogo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/fisiología
15.
Angew Chem Int Ed Engl ; 60(22): 12524-12531, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-33599016

RESUMEN

Among phosphorus-based nanomaterials, layered black phosphorus and violet phosphorus have been actively explored in the past decade. However, methods for the synthesis of red phosphorus nanosheets (RPNSs) is lacking, even though red phosphorus (RP) is commercially available at low cost and has excellent chemical stability at room temperature. We report an efficient strategy for fabrication of RPNSs and doped RPNSs using cysteine as a reducing reagent. Data from in vitro and in vivo studies suggested that RPNSs can trigger production of reactive oxygen species, DNA damage, and subsequent autophagy-mediated cell death in a shape-dependent manner. Our findings provide a method for construction of layered RP nanomaterials and they present a unique mechanism for the application of phosphorus-based materials in nanomedicines.


Asunto(s)
Cisteína/química , Nanoestructuras/química , Fósforo/química , Células A549 , Animales , Apoptosis/efectos de los fármacos , Compuestos de Boro/química , Línea Celular , Daño del ADN/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Nanoestructuras/uso terapéutico , Nanoestructuras/toxicidad , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Sustancias Reductoras/química , Trasplante Heterólogo
16.
Bioorg Chem ; 107: 104574, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33383327

RESUMEN

Small cell lung cancer (SCLC) is exceedingly tough to treat and easy to develop resistance upon long use of the first-line drug carboplatin or radiotherapy. Novel medicines effective and specific against SCLC are greatly needed. Herein, we focused on the discovery of such a medicine by exploring a drug niclosamide with repurposing strategy. Initial screening efforts revealed that niclosamide, an anthelmintic drug, possessed the in vitro anticancer activity and an obvious sensitivity towards SCLC. This observation inspired the evaluation for two different kinds of niclosamide derivatives. 2 with a degradable ester as a linker exhibited the comparable activity but slightly inferior selectivity to SCLC, by contrast, the cytotoxicities of 4 and 5 with non-degradable ether linkages completely disappeared, clearly validating the importance of 2-free hydroxyl group or 2-hydroxyl group released in the antitumor activity. Mechanism study unfolded that, similar to niclosamide, 2 inhibited growth of cancer cells via p 53 activation and subsequent underwent cytochrome c dependent apoptosis. Further structural modification to afford phosphate sodium 8 with significantly enhanced aqueous solubility (22.1 mg/mL) and a good selectivity towards SCLC demonstrated more promising druggability profiles. Accordingly, niclosamide as an attractive lead hold a huge potential for developing targeted anti-SCLC drugs.


Asunto(s)
Antineoplásicos/farmacología , Niclosamida/análogos & derivados , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Niclosamida/farmacología , Niclosamida/uso terapéutico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Solubilidad , Trasplante Heterólogo
17.
Acc Chem Res ; 54(3): 618-631, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33228351

RESUMEN

Here we provide a personal account of innovation and design principles underpinning a method to interrogate precision electrophile signaling that has come to be known as "REX technologies". This Account is framed in the context of trying to improve methods of target mining and understanding of individual target-ligand engagement by a specific natural electrophile and the ramifications of this labeling event in cells and organisms. We start by explaining from a practical standpoint why gleaning such understanding is critical: we are constantly assailed by a battery of electrophilic molecules that exist as a consequence of diet, food preparation, ineluctable endogenous metabolic processes, and potentially disease. The resulting molecules, which are detectable in the body, appear to be able to modify function of specific proteins. Aside from potentially being biologically relevant in their own right, these labeling events are essentially identical to protein-covalent drug interactions. Thus, on what proteins and even in what ways a covalent drug will work can be understood through the eyes of natural electrophiles; extending this logic leads to the postulate that target identification of specific electrophiles can inform on drug design. However, when we entered this field, there was no way to interrogate how a specific labeling event impacted a specific protein in an unperturbed cell. Methods to evaluate stoichiometry of labeling, and even chemospecificity of a specific phenotype were limited. There were further no generally accepted ways to study electrophile signaling that did not hugely disturb physiology.We developed T-REX, a method to study single-protein-specific electrophile engagement, to interrogate how single-protein electrophile labeling shapes pathway flux. Using T-REX, we discovered that labeling of several proteins by a specific electrophile, even at low occupancy, leads to biologically relevant signaling outputs. Further experimentation using T-REX showed that in some instances, single-protein isoforms were electrophile responsive against other isoforms, such as Akt3. Selective electrophile-labeling of Akt3 elicited inhibition of Akt-pathway flux in cells and in zebrafish embryos. Using these data, we rationally designed a molecule to selectively target Akt3. This was a fusion of the naturally derived electrophile and an isoform-nonspecific, reversible Akt inhibitor in phase-II trials, MK-2206. The resulting molecule was a selective inhibitor of Akt3 and was shown to fare better than MK-2206 in breast cancer xenograft mouse models. Recently, we have also developed a means to screen electrophile sensors that is unbiased and uses a precise burst of electrophiles. Using this method, dubbed G-REX, in conjunction with T-REX, we discovered new DNA-damage response upregulation pathways orchestrated by simple natural electrophiles. We thus emphasize how deriving a quantitative understanding of electrophile signaling that is linked to thorough and precise mechanistic studies can open doors to numerous medicinally and biologically relevant insights, from gleaning better understanding of target engagement and target mining to rational design of targeted covalent medicines.


Asunto(s)
Preparaciones Farmacéuticas/química , Proteínas Proto-Oncogénicas c-akt/química , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Portadores de Fármacos/química , Evaluación Preclínica de Medicamentos , Femenino , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/metabolismo , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Ligandos , Ratones , Oxidantes/química , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trasplante Heterólogo
18.
Cell Signal ; 77: 109819, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33147518

RESUMEN

Current treatment options of glioblastoma include chemotherapy and limited surgical resection. Temozolomide (TMZ) is the current therapeutic choice for chemotherapy. Still, it has severe limitations due to the development of resistance that occurs by genetic modification and constitutive activation of several cell signaling pathways. Therefore, it is essential to develop combination therapy of TMZ with other novel compounds to prevent the development of chemo-resistance. In this study, we used two inhibitors; ICA, an inhibitor of PKC-ι and ζ-Stat, an inhibitor of PKC-ζ. T98G and U87MG glioblastoma cells were treated with either ICA or ζ-stat or TMZ monotherapies, as well as TMZ were combined with either ICA or ζ-stat for five consecutive days. Our in vitro results exhibited that ICA when combined with TMZ, significantly decreased the viability of cancerous cells compared with untreated or TMZ or ICA monotherapies. Additionally, glioblastoma cells were remarkably undergoing apoptosis against the combination treatment of TMZ and ICA nucleotide compared with untreated control cells, as suggested by our Annexin-V/PI flow cytometric analysis. Moreover, the combination of TMZ and ICA also decreased the invasion of glioblastoma cell lines by acting on FAK/Paxillin pathway, as evidenced by scratch assay, transwell invasion assay, Western blot and immunoprecipitation analysis. Furthermore, our in vivo data presented that the combination of ICA and TMZ also reduced glioblastoma tumor growth and volume in mice. These data suggest that atypical PKCs, particularly PKC-ι might be an important therapeutic target as adjuvant therapy in the treatment of glioblastoma.


Asunto(s)
Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Temozolomida/farmacología , Citoesqueleto de Actina/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimioterapia Combinada , Quinasa 1 de Adhesión Focal/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Ratones , Ratones Desnudos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Temozolomida/uso terapéutico , Trasplante Heterólogo
19.
ACS Appl Mater Interfaces ; 12(49): 54399-54414, 2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33215918

RESUMEN

Tumor vaccines based on synthetic human papillomavirus (HPV) oncoprotein E7 and/or E6 peptides have shown encouraging results in preclinical model studies and human clinical trials. However, the clinical efficacy may be limited by the disadvantages of vulnerability to enzymatic degradation and low immunogenicity of peptides. To further improve the potency of vaccine, we developed a poly(lactide-co-glycolide)-acid (PLGA) nanoparticle, which encapsulated the antigenic peptide HPV16 E744-62, and used adenosine triphosphate (ATP), one of the most important intracellular metabolites and an endogenous extracellular danger signal for the immune system, as a new adjuvant component. The results showed that PLGA nanoparticles increased the in vivo stability, lymph node accumulation, and dendritic cell (DC) uptake of the E7 peptide; in addition, ATP further increased the migration, nanoparticle uptake, and maturation of DCs. Preventive immunization with ATP-adjuvanted nanoparticles completely abolished the growth of TC-1 tumors in mice and produced long-lasting immunity against tumor rechallenge. When tumors were fully established, therapeutic immunization with ATP-adjuvanted nanoparticles still significantly inhibited tumor progression. Mechanistically, ATP-adjuvanted nanoparticles significantly improved the systemic generation of antitumor effector cells, boosted the local functional status of these cells in tumors, and suppressed the generation and tumor infiltration of immunosuppressive Treg cells and myeloid-derived suppressor cells. These findings indicate that ATP is an effective vaccine adjuvant and that nanoparticles adjuvanted with ATP were able to elicit robust antitumor cellular immunity, which may provide a promising therapeutic vaccine candidate for the treatment of clinical malignancies, such as cervical cancer.


Asunto(s)
Adenosina Trifosfato/metabolismo , Vacunas contra el Cáncer/inmunología , Inmunidad Celular , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Adenosina Trifosfato/química , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Neoplasias/terapia , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/inmunología , Péptidos/química , Péptidos/inmunología , Péptidos/metabolismo , Trasplante Heterólogo
20.
Chem Commun (Camb) ; 56(78): 11589-11592, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32914792

RESUMEN

Alteration of the levels of copper is a promising approach for cancer therapy. Herein, we develop a dual-mode copper vehicle, M985. The biotin-tailed M985 can exert tumor-directed copper supplementation and undergo self-immolative cleavage in living cancerous cells, resulting in the liberation of F542 along with the generation of excess reactive oxygen species. Thus, fluorescence and 19F NMR detection is realized to specifically discriminate cancer cells. F542 acts as a fluorescence reporter and a potent cytotoxic agent, facilitating the visualization of molecular release and distribution, as well as confirming the ER autophagy-induced apoptosis. Therefore, we present a promising dual-mode theranostic M985 for the efficient detection and therapy of cancer.


Asunto(s)
Complejos de Coordinación/química , Cobre/química , Espectroscopía de Resonancia Magnética , Neoplasias/diagnóstico por imagen , Nanomedicina Teranóstica , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Biotina/química , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Retículo Endoplásmico/metabolismo , Flúor/química , Glutatión/química , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Imagen Óptica , Especies Reactivas de Oxígeno/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA