Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 15: 1396133, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38828460

RESUMO

Objective: Growth differentiation factor 15 (GDF-15) is a stress-responsive cytokine that regulates myocardial injury, cardiac overloading pressure, and inflammation and is related to the risk of cardiovascular diseases and events. The current study aimed to investigate the correlation of GDF-15 levels with clinical features, biochemical indices, and especially the risk of cardiotoxicity in breast cancer patients receiving neoadjuvant dual anti-HER2 therapy. Methods: A total of 103 HER2-positive breast cancer patients who underwent neoadjuvant dual anti-HER2 therapy (trastuzumab and pertuzumab plus chemotherapy) were included. Serum GDF-15 levels before neoadjuvant treatment were detected by enzyme-linked immunosorbent assay. Cardiotoxicity was evaluated during neoadjuvant therapy by referring to a decline of ≥10 percentage points in the left ventricular ejection fraction from baseline to an absolute level less than 50%. Results: GDF-15 exhibited a skewed distribution, with a median level of 714 (range: 207-1805) pg/mL. GDF-15 was positively correlated with age (p = 0.037), diabetes (p = 0.036), and the N-terminal pro-brain natriuretic peptide level (p = 0.013) and positively correlated with the total cholesterol level (p = 0.086) and troponin T level (p = 0.082), but these correlations were not statistically significant. A total of 6.8% of patients experienced cardiotoxicity during neoadjuvant therapy. By comparison, the GDF-15 level was greater in patients who experienced cardiotoxicity than in those who did not (p = 0.008). A subsequent receiver operating characteristic curve revealed that GDF-15 predicted cardiotoxicity risk, with an area under the curve of 0.803 (95% CI: 0.664-0.939). After multivariate adjustment, GDF-15 independently predicted a greater risk of cardiotoxicity (p = 0.020). Conclusion: GDF-15 is a candidate biomarker for increased risk of cardiotoxicity in breast cancer patients receiving neoadjuvant dual anti-HER2 therapy.

2.
Pharmaceutics ; 15(8)2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37631241

RESUMO

Photothermal therapy operated in the second near-infrared (NIR-II, 1000-1700 nm) window and fluorescence imaging in the NIR-IIb (1500-1700 nm) region have become the most promising techniques in phototheranostics. Their combination enables simultaneous high-resolution optical imaging and deep-penetrating phototherapy, which is essential for high-performance phototheranostics. Herein, carboxyl-functionalized small organic photothermal molecules (Se-TC) and multi-layered NIR-IIb emissive rare-earth-doped nanoparticles (NaYF4:Yb,Er,Ce@NaYF4:Yb,Nd@NaYF4, RENP) were rationally designed and successfully synthesized. Then, high-performance hybrid phototheranostic nanoagents (Se-TC@RENP@F) were easily constructed through the coordination between Se-TC and RENP and followed by subsequent F127 encapsulation. The carboxyl groups of Se-TC can offer strong binding affinity towards rare-earth-doped nanoparticles, which help improving the stability of Se-TC@RENP@F. The multilayered structure of RENP largely enhance the NIR-IIb emission under 808 nm excitation. The obtained Se-TC@RENP@F exhibited high 1064 nm absorption (extinction coefficient: 24.7 L g-1 cm-1), large photothermal conversion efficiency (PCE, 36.9%), good NIR-IIb emission (peak: 1545 nm), as well as great photostability. Upon 1064 nm laser irradiation, high hyperthermia can be achieved to kill tumor cells efficiently. In addition, based on the excellent NIR-IIb emission of Se-TC@RENP@F, in vivo angiography and tumor detection can be realized. This work provides a distinguished paradigm for NIR-IIb-imaging-guided NIR-II photothermal therapy and establishes an artful strategy for high-performance phototheranostics.

3.
Semin Cancer Biol ; 94: 62-80, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37302519

RESUMO

The use of artificial intelligence (AI) to assist biomedical imaging have demonstrated its high accuracy and high efficiency in medical decision-making for individualized cancer medicine. In particular, optical imaging methods are able to visualize both the structural and functional information of tumors tissues with high contrast, low cost, and noninvasive property. However, no systematic work has been performed to inspect the recent advances on AI-aided optical imaging for cancer theranostics. In this review, we demonstrated how AI can guide optical imaging methods to improve the accuracy on tumor detection, automated analysis and prediction of its histopathological section, its monitoring during treatment, and its prognosis by using computer vision, deep learning and natural language processing. By contrast, the optical imaging techniques involved mainly consisted of various tomography and microscopy imaging methods such as optical endoscopy imaging, optical coherence tomography, photoacoustic imaging, diffuse optical tomography, optical microscopy imaging, Raman imaging, and fluorescent imaging. Meanwhile, existing problems, possible challenges and future prospects for AI-aided optical imaging protocol for cancer theranostics were also discussed. It is expected that the present work can open a new avenue for precision oncology by using AI and optical imaging tools.


Assuntos
Inteligência Artificial , Neoplasias , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Medicina de Precisão , Tomografia de Coerência Óptica/métodos , Oncologia
4.
Ir J Med Sci ; 192(3): 1041-1049, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35829909

RESUMO

OBJECTIVE: Pyrotinib is a novel EGFR/HER2 dual tyrosine kinase inhibitor developed in China, while its role in neoadjuvant therapy of HER2-positive (HER2+) breast cancer lacks evidence. The current study aimed to explore the efficacy and safety of neoadjuvant pyrotinib plus docetaxel/liposomal doxorubicin/cyclophosphamide (TAC) for HER2+ breast cancer. METHODS: A total of 27 HER2+ breast cancer patients received neoadjuvant pyrotinib plus TAC for 6 cycles, then surgery was performed. The clinical and pathological responses, and adverse events were evaluated. RESULTS: Complete response rate, objective response rate, and disease control rate were 0.0%, 44.4% and 100.0% after 2 treatment cycles; 0.0%, 37.0%, and 100.0% after 4 treatment cycles; 37.0%, 37.0%, and 96.3% after 6 treatment cycles; as well as 37.0%, 44.4%, and 100.0% based on the best clinical response. Regarding pathological response, there were 1 (2.7%), 3 (11.1%), 8 (29.6%), 5 (18.5%), and 10 (37.0%) patients realizing Miller-Payne grade (G) 1, G2, G3, G4, and G5, respectively; besides, 10 (37.0%) patients achieved total pathological complete response (pCR), 10 (37.0%) patients realized pCR in breast, and 23 (85.2%) patients achieved pCR in lymph node. Additionally, adverse events included diarrhea (81.5%), dental ulcer (7.4%), and hand-foot syndrome (3.7%); meanwhile, grade 3-4 adverse event consisted of only diarrhea (11.1%). CONCLUSION: Neoadjuvant pyrotinib plus TAC treatment is efficient and safe in HER2+ breast cancer patients, while further validation is needed.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/cirurgia , Docetaxel/uso terapêutico , Terapia Neoadjuvante , Ciclofosfamida/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Receptor ErbB-2
5.
Pharmaceutics ; 14(7)2022 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-35890265

RESUMO

Photoacoustic imaging and photothermal therapy that employ organic dye in the second near-infrared window (NIR-II) became an attractive theranostical strategy for eliminating solid tumors, in which IR1048 was previously reported to be a good candidate. However, the further biomedical application of IR1048 was blocked by its poor water-solubility and lack of tumor-targeting. To solve this problem, liposome camouflaged with 4T1 cell membrane fragments was employed to encapsulate IR1048 (thereafter called MLI), and its application for photoacoustic and thermo-imaging and photothermal therapy were explored in vitro and in vivo. The results showed that MLI exhibited spherical morphology around 92.55 ± 5.41 nm coated by monolayer adventitial fragments, and uniformly dispersed in PBS with high loading efficiency and encapsulation efficiency to IR1048. In addition, both free IR1048 and MLI presented strong absorption in NIR-II, and upon 1064 nm laser irradiation the MLI showed awesome photothermal performance that could rapidly elevate the temperature to 50.9 °C in 6 min. Simultaneously, phantom assay proved that MLI could dramatically enhance the photoacoustic amplitudes by a linear concentration-dependent way. Moreover, either flow cytometry or confocal analysis evidenced that MLI was the most uptaked by 4T1 cells among other melanoma B16 cells and Hek293 cells and coexist of IR1048 and 1064 nm laser irradiation were indispensable for the photothermal cytotoxicity of MLI that specifically killed 96.16% of 4T1 cells far outweigh the B16 cells while hardly toxic to the Hek293 normal cells. Furthermore, PA imaging figured out that 4 h post tail-vein injection of MLI was the best time to give 1064 nm irradiation to conduct the photothermal therapy when the average tumor-accumulation of MLI achieved the highest. In the NIR-II photothermal therapy, MLI could significantly inhibit the tumor growth and almost ablated the tumors with slight body weight variation and the highest average life span over the therapy episode and caused no damage to the normal organs. Hence, MLI could pave the way for further biomedical applications of IR-1048 by homologous tumor-targeting and dual-modal imaging directed NIR-II accurate photothermal therapy with high efficacy and fine biosafety.

6.
J Am Chem Soc ; 144(2): 787-797, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-34985903

RESUMO

Tumor-derived exosome can suppress dendritic cells (DCs) and T cells functions. Excessive secretion of exosomal programmed death-ligand 1 (PD-L1) results in therapeutic resistance to PD-1/PD-L1 immunotherapy and clinical failure. Restored T cells by antiexosomal PD-L1 tactic can intensify ferroptosis of tumor cells and vice versa. Diminishing exosomal suppression and establishing a nexus of antiexosomal PD-L1 and ferroptosis may rescue the discouraging antitumor immunity. Here, we engineered phototheranostic metal-phenolic networks (PFG MPNs) by an assembly of semiconductor polymers encapsulating ferroptosis inducer (Fe3+) and exosome inhibitor (GW4869). The PFG MPNs elicited superior near-infrared II fluorescence/photoacoustic imaging tracking performance for a precise photothermal therapy (PTT). PTT-augmented immunogenic cell death relieved exosomal silencing on DC maturation. GW4869 mediated PD-L1 based exosomal inhibition revitalized T cells and enhanced the ferroptosis. This novel synergy of PTT with antiexosomal PD-L1 enhanced ferroptosis evoked potent antitumor immunity in B16F10 tumors and immunological memory against metastatic tumors in lymph nodes.


Assuntos
Compostos de Anilina/química , Antígeno B7-H1/metabolismo , Compostos de Benzilideno/química , Compostos Férricos/química , Ferroptose , Estruturas Metalorgânicas/química , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Exossomos/metabolismo , Ferroptose/efeitos dos fármacos , Morte Celular Imunogênica/efeitos dos fármacos , Imunoterapia , Interferon gama/metabolismo , Melanoma Experimental/diagnóstico por imagem , Melanoma Experimental/terapia , Estruturas Metalorgânicas/farmacologia , Estruturas Metalorgânicas/uso terapêutico , Camundongos , Fenol/química , Técnicas Fotoacústicas , Polietilenoglicóis/química , Polímeros/química , Receptor de Morte Celular Programada 1/metabolismo , Nanomedicina Teranóstica
7.
Nanoscale ; 13(31): 13410-13420, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34477746

RESUMO

Photoacoustic imaging (PAI)-guided photothermal therapy (PTT) has drawn considerable attention due to the deeper tissue penetration and higher maximum permissible exposure. However, current phototheranostic agents are greatly restricted by weak absorption in the second near-infrared (NIR-II, 1000-1700 nm) window, long-term toxicity, and poor photostability. In this report, novel organic NIR-II conjugated polymer nanoparticles (CPNs) based on narrow bandgap donor-acceptor BDT-TBZ polymers were developed for effective cancer PAI and PTT. Characterization data confirmed the high photothermal conversion efficiency, good photostability, excellent PAI performance, and superior biocompatibility of as-obtained CPNs. In addition, in vitro and in vivo tests demonstrated the efficient PTT effect of CPNs in ablating cancer cells and inhibiting tumor growth under 1064 nm laser irradiation. More importantly, the CPNs exhibited rapid clearance capability through the biliary pathway and negligible systematic toxicity. Thus, this work provides a novel organic theranostic nanoplatform for NIR-II PAI-guided PTT, which advances the future clinical translation of biocompatible and metabolizable conjugated nanomaterials in cancer diagnosis and therapy.


Assuntos
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Fototerapia , Polímeros , Medicina de Precisão , Nanomedicina Teranóstica
8.
Small ; 17(43): e2101332, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34405525

RESUMO

Inorganic nanomedicine has attracted increasing attentions in biomedical sciences due to their excellent biocompatibility and tunable, versatile functionality. However, the relatively poor accumulation and retention of these nanomedicines in targeted tissues have often hindered their clinical translation. Herein, highly efficient, targeted delivery, and in situ aggregation of ferrocene (Fc)-capped Au nanoparticles (NPs) are reported to cucurbit[7]uril (CB[7])-capped Fe3 O4 NPs (as an artificial target) that are magnetically deposited into the tumor, driven by strong, multipoint CB[7]-Fc host-guest interactions (here defined as "supramolecular tropism" for the first time), leading to high tumor accumulation and retention of these NPs. The in vitro and in vivo studies demonstrate the precisely controlled, specific accumulation, and retention of Au NPs in the tumor cells and tissue via supramolecular tropism and in situ aggregation, which afford locally enhanced CT imaging of cancer and enable tumor-specific photothermal therapy attributed to the plasmonic coupling effects between adjacent Au NPs within the supramolecular aggregations. This work provides a novel concept of supramolecular tropism, which may drive targeted delivery and enable specific accumulation, retention, and activation of nanomedicine for improved bioimaging and therapy of cancer.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias , Ouro , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Terapia Fototérmica , Tropismo
9.
Small ; 17(43): e2101397, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34159726

RESUMO

In this study, to visually acquire all-round structural and functional information of lung cancer while performing synergistic photothermal therapy (PTT) and tumor-targeting immunotherapy, a theranostic nanoplatform that introduced upconversion nanoparticles (UCNPs) and IR-1048 dye into the lipid-aptamer nanostructrure (UCILA) is constructed. Interestingly, the IR-1048 dye grafted into the lipid bilayer can serve as the theranostic agent for photoacoustic imaging, optical coherence tomography angiography, photothermal imaging, and PTT in the second near infrared (NIR-II) window. In addition, loaded in the inner part of UCILA, UCNPs possess the superior luminescence property and high X-ray attenuation coefficient, which can act as contrast agents for computed tomography (CT) and thermo-sensitive up-conversion luminescence (UCL) imaging, enabling real-time tracking of metabolic activity of tumor and temperature-feedback PTT. Furthermore, under the complementary guidance of penta-modal imaging and an accurate monitoring of in situ temperature change during PTT, UCILA exhibits its excellent capability for ablating the lung tumor with minimal side effects. Meanwhile, synergistic CAR-NK immunotherapy is carried out specifically to eradicate any possible residual tumor cells after PTT. Therefore, the UCILA nanoplatform is demonstrated as a multifunctional theranostic agent for both penta-modal imaging and temperature-feedback PTT while conducting targeting immunotherapy of lung cancer.


Assuntos
Hipertermia Induzida , Neoplasias Pulmonares , Nanopartículas , Rubiaceae , Linhagem Celular Tumoral , Retroalimentação , Humanos , Imunoterapia , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/terapia , Fototerapia , Terapia Fototérmica , Temperatura , Nanomedicina Teranóstica
10.
Biomaterials ; 275: 120822, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34062419

RESUMO

The hypoxic tumor microenvironment (TME) and non-specific distribution of sonosensitizers are two major obstacles that limit practical applications of sonodynamic therapy (SDT) in combating tumors. Here we report a hypoxia-responsive nanovesicle (hMVs) as delivery vehicles of a sonosensitizer to enhance the efficacy of SDT via specific payload release and local oxygenation in the tumor. The nanovesicles are composed of densely packed manganese ferrite nanoparticles (MFNs) embedded in hypoxia-responsive amphiphilic polymer membranes. With δ-aminolevulinic acid (ALA) loaded in the hollow cavities, the hMVs could rapidly dissociate into discrete nanoparticles in the hypoxic TME to release the payload and induce the generation of reactive oxygen species (ROS) under ultrasound (US) radiation. Meanwhile, the released MFNs could catalytically generate O2 to overcome the hypoxic TME and thus enhance the efficacy of SDT. After treatment, the dissociated MFNs could be readily excreted from the body via renal clearance to reduce long term toxicity. In vitro and in vivo experiments displayed effective tumor inhibition via hMVs-mediated SDT, indicating the great potential of this unique nanoplatform in effective SDT by generating sufficient ROS in deep-seated hypoxic tumors that are not readily accessible by conventional photodynamic therapy.


Assuntos
Hipóxia , Nanopartículas , Linhagem Celular Tumoral , Humanos , Espécies Reativas de Oxigênio , Microambiente Tumoral
11.
Biomaterials ; 251: 120088, 2020 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-32388167

RESUMO

Hypoxia is one of the hallmarks of solid tumor, which heavily restricts the clinical cancer therapy treatments, especially for the oxygen (O2) -dependent photodynamic therapy (PDT). Herein, an intelligent multi-layer nanostructure was developed for decreasing the O2-consumption and elevating the O2-supply simultaneously. The cell respiration inhibitor -atovaquone (ATO) molecules were reserved in the middle mesoporous silicon layer, and thus were intelligently released at the tumor site after the degradation of gatekeeper of MnO2 layer, which effectively inhibit tumor respiration metabolism to elevate oxygen content. Meanwhile, the degradation of MnO2 layer can generate O2, further boosting oxygen content. Moreover, the inner upconversion nanostructures as the near infrared (NIR) light-transducers enable to activate photosensitizers for deep-tissue PDT. Systematic experiments demonstrate that this suppressing O2-consumption and O2-generation strategy improved oxygen supply to boost the singlet oxygen generation to eradicate cancer cells under NIR light excitation. Better still, superior trimodality imaging capabilities (computed tomography (CT), NIR-II window fluorescence, and tumor microenvironment-responsive T1-weighted magnetic resonance (MR) imaging) of the nanoplatform were evaluated. Our findings offer a promising aproach to conquer the serious hypoxia problem in cancer therapy by turning down the O2 metabolism aveneue and simultaneously generating O2.

12.
Chem Commun (Camb) ; 55(44): 6209-6212, 2019 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-31073580

RESUMO

In this study, pH-sensitive loaded retinal/indocyanine green (ICG) micelles were developed to realize novel approaches for cellular senescence-photothermal synergistic therapy to treat cancer. The micelles could enable effective multi-modal imaging in vivo guided therapy and show anticancer activity in vitro and in vivo with satisfactory biosafety.


Assuntos
Senescência Celular , Concentração de Íons de Hidrogênio , Verde de Indocianina/metabolismo , Micelas , Imagem Multimodal , Fototerapia/métodos , Retinaldeído/metabolismo , Nanomedicina Teranóstica , Humanos , Neoplasias/patologia
13.
J Biophotonics ; 12(3): e201800237, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30414259

RESUMO

In this study, CuS nanoparticles with optical absorption covering both near-infrared I (NIR-I) and NIR-II biological windows were prepared and served as the contrast agents for multispectral photoacoustic imaging. The physiological parameters including concentrations of deoxyhemoglobin and oxyhemoglobin as well as the water content in the tumor location were quantified based on the multispectral photoacoustic reconstruction method. More importantly, the concentration of CuS nanoparticles/drugs accumulated in the tumor was also recovered after intravenously injection, which are essential for image-guided cancer theranostics. In addition, phantom and in vivo experimental tests were performed to inspect and compare the imaging depth and signal-to-noise ratio (SNR) between the two NIR biological windows. Interestingly, we discovered that a higher SNR was obtained in the NIR-II window than that in the NIR-I window. Meanwhile, the multispectral imaging results also demonstrated that the imaging contrast and penetration depth in the NIR-II window were also significantly improved as compared to those from the NIR-I window.


Assuntos
Cobre/química , Raios Infravermelhos , Nanopartículas/química , Técnicas Fotoacústicas/métodos , Tomografia/métodos , Linhagem Celular Tumoral , Meios de Contraste/química , Células HEK293 , Humanos , Processamento de Imagem Assistida por Computador , Imagens de Fantasmas , Razão Sinal-Ruído
14.
Biomater Sci ; 5(12): 2501-2510, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29119997

RESUMO

Camptothecin (CPT) is a broad spectrum anticancer drug, but its application is limited due to the poor water solubility, lactone ring instability, and low drug loading potential. In this study, biocompatible cationic polypeptide-based micelles were developed to deliver dimeric CPT (DCPT) with the aim of overcoming the above-mentioned obstacles and achieving favorable therapeutic effects. Cationic polypeptide poly-lysine-block-poly-leucine (PLys-b-PLeu) was fabricated via the ring-opening polymerization of N-ε-carbobenzoxy-l-lysine (ε-Lys(Z)) and l-leucine (Leu) and further grafted with polyethylene glycol (PEG) and an arginine-glycine-aspartic acid (RGD) peptide. DCPT was synthesized by reacting CPT and 2-hydroxyethyl disulfide, and micelles were prepared using a dialysis method. The obtained DCPT-loaded RGD-PEG-g-poly-l-lysine-b-poly-l-leucine (DRPPP) micelles showed a high encapsulation efficiency of 89.7% and a high drug loading capacity of 46.1%. In addition, the DRPPP micelles remained stable under physiological conditions (PBS at a pH of 7.4) but showed rapid release when triggered by a reductive environment (PBS at a pH of 7.4 with 10 mM dithiothreitol). Compared to micelles without RGD decoration, the DRPPP micelles exhibited an increased cellular uptake through RGD targeting and were internalized into cells via caveolae-mediated endocytosis and macropinocytosis. Furthermore, the DRPPP micelles exerted an enhanced cytotoxicity against MDA-MB-231 cells compared to MCF-7 cells, which expressed less αvß3 receptors. Besides, the DRPPP micelles induced cell apoptosis and caused a decrease of mitochondrial membrane potential. These results indicate that dimeric camptothecin-loaded cationic polypeptide-based micelle is a promising strategy for cancer therapy.


Assuntos
Camptotecina/administração & dosagem , Liberação Controlada de Fármacos , Neoplasias/tratamento farmacológico , Peptídeos/química , Camptotecina/química , Cátions/administração & dosagem , Cátions/química , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Humanos , Células MCF-7 , Micelas , Neoplasias/patologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/química , Oxirredução/efeitos dos fármacos , Peptídeos/administração & dosagem , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química
15.
Bioorg Med Chem ; 22(24): 6746-54, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25468038

RESUMO

A series of 7,8-dihydro-5H-thiopyrano[4,3-d]pyrimidine derivatives (7a-q, 10a-q) were designed, synthesized and their chemical structures were confirmed by 1H NMR, 13C NMR, MS and HRMS spectrum.All the compounds were evaluated for the inhibitory activity against mTOR kinase at 10 µM level. Five selected compounds (7b, 7e, 7h, 10b and 10e) were further evaluated for the inhibitory activity against PI3Ka at 10 µM level, and the IC50 values against mTOR kinase and two cancer cell lines. Twelve of the target compounds exhibited moderate antitumor activities. The most promising compound 7e showed strong antitumor activities against mTOR kinase, H460 and PC-3 cell lines with IC50 values of 0.80 ± 0.15 µM, 7.43 ± 1.45 µM and 11.90 ± 0.94 µM, which were 1.28 to 1.71-fold more active than BMCL-200908069-1 (1.37 ± 0.07 µM, 9.52 ± 0.29 µM, 16.27 ± 0.54 µM), respectively. Structure-activity relationships (SARs) and docking studies indicated that the thiopyrano[4,3-d]pyrimidine scaffolds exerted little effect on antitumor activities of target compounds. Substitutions of aryl group at C-4 position had a significant impact on the antitumor activities, and 4-OH substitution produced the best potency.


Assuntos
Antineoplásicos/síntese química , Desenho de Fármacos , Inibidores de Proteínas Quinases/síntese química , Pirimidinas/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/toxicidade , Sítios de Ligação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/toxicidade , Estrutura Terciária de Proteína , Pirimidinas/síntese química , Pirimidinas/toxicidade , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA