Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
1.
BMC Complement Med Ther ; 21(1): 133, 2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33926427

RESUMO

BACKGROUND: Cardamom (Elettaria cardamomum) is a spice and exhibits potent antioxidant and biological activities through distinct molecular mechanisms. However, the anticancer effect of cardamom was not explored yet in Ehrlich solid tumor (EST)-bearing mice. OBJECTIVES: This investigation was aimed to evaluate the anti-cancer effects of green cardamom (GCar) alone or combined with the anti-cancer drug cyclophosphamide in an in vivo model to explore its mechanistic role in tumor cell death in EST-bearing mice. METHODS: Ehrlich ascites tumor cells were injected in the mice and 5 days later the animals treated with GCar and/or cyclophosphamide for 10 days. Twenty-four hours from the last treatment, animals were sacrificed for the different measurements. RESULTS: Data recorded for tumor size, percentage of tumor growth inhibition, tumor growth delay and mean survival time of EST-bearing mice demonstrated the effective role of GCar alone or combined with CPO as a promising anti-cancer agent because it reduced tumor size. GCar elevated the mean survival time of EST-bearing mice compared to that of untreated EST and EST + CPO groups. Analysis of qPCR mRNA gene and protein expression revealed that GCar alone or combined with CPO were promising anticancer agents. After the treatment of EST with GCar, the apoptotic-related genes and proteins were significantly modulated. GCar induced markedly significant decreases in oxidative stress biomarkers and a significant increment in glutathione levels and that of antioxidant enzymes. With a marked diminish in liver and kidney function biomarkers. CONCLUSION: The results revealed that GCar could serve as an apoptotic stimulator agent, presenting a novel and potentially curative approach for cancer treatment, inducing fewer side effects than those of the commercially used anti-cancer drugs, such as CPO.


Assuntos
Antineoplásicos , Carcinoma de Ehrlich , Ciclofosfamida , Elettaria , Extratos Vegetais , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Peso Corporal/efeitos dos fármacos , Carcinoma de Ehrlich/química , Carcinoma de Ehrlich/patologia , Ciclofosfamida/farmacologia , Ciclofosfamida/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/química , Neoplasias Experimentais/patologia , Extratos Vegetais/farmacologia , Extratos Vegetais/toxicidade , Sementes/química
2.
ACS Appl Bio Mater ; 4(3): 2001-2008, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35014325

RESUMO

Cysteine (Cys) is well-known to be an important biothiol and related to many diseases. However, the in situ trapping of endogenous Cys is still handicapped by a lack of straightforward methods combined with long-wavelength emission and high-performance response. In this work, we described the rational design strategy of cyanine-based near-infrared (NIR) probes for the rapid detection of mitochondrial Cys in living cells and mice. We focus on how to improve the response rate via regulating the electron density of the recognition units in probes. The obtained three probes all displayed remarkable fluorescence enhancement at 780 nm. From screening the obtained probes, it was found that the probe Cy-S-diOMe with electron-donating recognition unit displayed the fastest response rate, the lowest detection limit, and the highest signal-to-noise ratio. More importantly, Cy-S-diOMe was successfully applied to monitor Cys in tumor-bearing mice (within merely 5 min). This paradigm by modulation of the response rate in the cyanine dyes provides a promising methodology for the design of high-performance cyanine-based NIR probes.


Assuntos
Materiais Biocompatíveis/química , Cisteína/análise , Desenho de Fármacos , Corantes Fluorescentes/química , Animais , Materiais Biocompatíveis/síntese química , Corantes Fluorescentes/síntese química , Células HeLa , Humanos , Raios Infravermelhos , Teste de Materiais , Camundongos , Estrutura Molecular , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Tamanho da Partícula
3.
Phytother Res ; 35(1): 504-516, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32869401

RESUMO

Breast cancer is one of the most lethal types of cancer and a leading cause of mortality among Women worldwide. Citrinin (CIT), a polyketide extracted from the fungus Penicillium citrinum, exhibits a wide range of biological activities such as antibacterial, antifungal, and cytotoxic effects. The aim of the current study was to evaluate the antitumoral effects of CIT against 7,12-dimethylbenzanthracene (DMBA)-induced mammary carcinoma in Swiss mice For this, CIT, DMBA and the standard cyclophosphamide (CPA) induced behavioral changes in experimental animals, and these changes were screened by using the rota rod and open field tests. Additionally, hematological, biochemical, immuno-histochemical, and histopathological analyses were carried out. Results suggest that CIT did not alter behavioral, hematological, and biochemical parameters in mice. DMBA induced invasive mammary carcinoma and showed genotoxic effects in the breasts, bone marrow, lymphocytes, and hepatic cells. It also caused mutagenic effects in the formation of micronuclei, bridges, shoots, and binucleate cells in bone marrow and liver. CIT and CPA genotoxic effects were observed after 3 weeks of therapy, where CIT exhibited a repair capacity and induced significant apoptotic damage in mouse lymphocytes. In conclusion, CIT showed antitumoral effects in Swiss mice, possibly through induction of apoptosis.


Assuntos
Antineoplásicos/farmacologia , Citrinina/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Penicillium/química , 9,10-Dimetil-1,2-benzantraceno , Animais , Apoptose/efeitos dos fármacos , Ciclofosfamida/farmacologia , Dano ao DNA/efeitos dos fármacos , Feminino , Camundongos , Mutagênicos , Neoplasias Experimentais/química
4.
Sci Rep ; 10(1): 15535, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968211

RESUMO

Penetration of nanoparticles into viable tumor regions is essential for an effective response. Mass spectrometry imaging (MSI) is a novel method for evaluating the intratumoral pharmacokinetics (PK) of a drug in terms of spatial distribution. The application of MSI for analysis of nanomedicine PK remains in its infancy. In this study, we evaluated the applicability of MALDI-MSI for nanoparticle-formulated drug visualization in tumors and biopsies, with an aim toward future application in clinical nanomedicine research. We established an analytic method for the free drug (AZD2811) and then applied it to visualize nanoparticle-formulated AZD2811. MSI analysis demonstrated heterogeneous intratumoral drug distribution in three xenograft tumors. The intensity of MSI signals correlated well with total drug concentration in tumors, indicating that drug distribution can be monitored quantitatively. Analysis of tumor biopsies indicated that MSI is applicable for analyzing the distribution of nanoparticle-formulated drugs in tumor biopsies, suggesting clinical applicability.


Assuntos
Acetanilidas/análise , Antineoplásicos/análise , Nanopartículas/análise , Neoplasias/química , Quinazolinas/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Acetanilidas/farmacocinética , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos/análise , Portadores de Fármacos/farmacocinética , Masculino , Neoplasias Mamárias Experimentais/química , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Experimentais/química , Quinazolinas/farmacocinética
6.
Angew Chem Int Ed Engl ; 59(48): 21546-21552, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32729159

RESUMO

Radiation-induced cleavage for controlled release in vivo is yet to be established. We demonstrate the use of 3,5-dihydroxybenzyl carbamate (DHBC) as a masking group that is selectively and efficiently removed by external radiation in vitro and in vivo. DHBC reacts mainly with hydroxyl radicals produced by radiation to afford hydroxylation at para/ortho positions, followed by 1,4- or 1,6-elimination to rescue the functionality of the client molecule. The reaction is rapid and can liberate functional molecules under physiological conditions. This controlled-release platform is compatible with living systems, as demonstrated by the release of a rhodol fluorophore derivative in cells and tumor xenografts. The combined benefits of the robust caging group, the good release yield, and the independence of penetration depth make DHBC derivatives attractive chemical caging moieties for use in chemical biology and prodrug activation.


Assuntos
Corantes Fluorescentes/metabolismo , Radical Hidroxila/metabolismo , Neoplasias/metabolismo , Xantonas/metabolismo , Animais , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Humanos , Radical Hidroxila/química , Hidroxilação , Camundongos , Estrutura Molecular , Neoplasias/química , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Xantonas/química
7.
Cancer Prev Res (Phila) ; 13(2): 185-194, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31699708

RESUMO

Recent observational studies suggest that bisphosphonates (BP) and antidiabetic drugs are associated with colorectal cancer risk reduction. Hence, we evaluated the colorectal cancer preventive effects of BPs (zometa and fosamax), individually and when combined with metformin, in azoxymethane-induced rat colon cancer model. Rat (30/group) were randomized and treated subcutaneously with azoxymethane to induce colorectal cancer. Dietary intervention with zometa or fosamax (0, 20, or 100 ppm) or metformin (1,000 ppm) or the combinations (zometa/fosamax 20 ppm plus metformin 1,000 ppm) began 4 weeks after azoxymethane treatment, at premalignant lesions stage. Rats were killed 40 weeks post drug intervention to assess colorectal cancer preventive efficacy. Dietary zometa (20 ppm) inhibited noninvasive adenocarcinomas multiplicity by 37% (P < 0.03) when compared with control diet fed group. Fosamax at 20 ppm and 100 ppm significantly reduced adenocarcinoma incidence (P < 0.005) and inhibited the noninvasive adenocarcinoma multiplicities by 43.8% (P < 0.009) and 60.8% (P < 0.004), respectively, compared with the group fed control diet. At 1,000 ppm dose, metformin failed to suppress colon adenocarcinoma formation. However, the lower dose combinations of zometa or fosamax with metformin resulted in significant inhibition of noninvasive adenocarcinoma by 48% (P < 0.006) and 64% (P < 0.0002), and invasive adenocarcinoma by 49% (P < 0.0005) and 38% (P < 0.006), respectively. Biomarker analysis of combination drug-treated tumors showed a decrease in cell proliferation with increased apoptosis when compared with untreated tumors. Overall, our results suggest that the combination of low doses of zometa or fosamax with metformin showed synergistic effect and significantly inhibited colon adenocarcinoma incidence and multiplicity.


Assuntos
Alendronato/farmacologia , Anticarcinógenos/farmacologia , Neoplasias do Colo/prevenção & controle , Metformina/farmacologia , Neoplasias Experimentais/prevenção & controle , Ácido Zoledrônico/farmacologia , Administração Oral , Alendronato/uso terapêutico , Animais , Anticarcinógenos/uso terapêutico , Apoptose/efeitos dos fármacos , Azoximetano/toxicidade , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Masculino , Metformina/uso terapêutico , Neoplasias Experimentais/química , Ratos , Ratos Endogâmicos F344 , Ácido Zoledrônico/uso terapêutico
8.
Anticancer Agents Med Chem ; 19(18): 2269-2275, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31692439

RESUMO

BACKGROUND: As involved in tumor angiogenesis, Neuropilin Receptor type-1 (NRP-1) serves as an attractive target for cancer molecular imaging and therapy. Widespread expression of NRP-1 in normal tissues may affect anti-NRP-1 antibody tumor uptake. OBJECTIVE: To assess a novel anti-NRP-1 monoclonal antibody A6-11-26 biodistribution in NRP-1 positive tumor xenograft models to understand the relationships between dose, normal tissue uptake and tumor uptake. METHODS: The A6-11-26 was radiolabeled with 131I and the mice bearing U87MG xenografts were then administered with 131I-labelled A6-11-26 along with 0, 2.5, 5, and 10mg·kg-1 unlabelled antibody A6-11-26. Biodistribution and SPECT/CT imaging were evaluated. RESULTS: 131I-A6-11-26 was synthesized successfully by hybridoma within 60min. It showed that most of 131IA6- 11-26 were in the plasma and serum (98.5 ± 0.16 and 88.9 ± 5.84, respectively), whereas, less in blood cells. For in vivo biodistribution studies, after only injection of 131I-A6-11-26, high levels of radioactivity were observed in the liver, moderate level in lungs. However, liver and lungs radioactivity uptakes could be competitively blocked by an increasing amount of unlabeled antibody A6-11-26, which can increase tumor radioactivity levels, but not in a dose-dependent manner. A dose between 10 and 20mg·kg-1 of unlabeled antibody A6-11-26 may be the optimal dose that maximized tumor exposure. CONCLUSION: Widespread expression of NRP-1 in normal tissue may affect the distribution of A6-11-26 to tumor tissue. An appropriate antibody A6-11-26 dose would be required to saturate normal tissue antigenic sinks to achieve acceptable tumor exposure.


Assuntos
Anticorpos Monoclonais/farmacocinética , Glioma/química , Neuropilina-1/genética , Compostos Radiofarmacêuticos/farmacocinética , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Glioma/metabolismo , Humanos , Radioisótopos do Iodo , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Neuropilina-1/metabolismo , Compostos Radiofarmacêuticos/química , Distribuição Tecidual
9.
Chem Biodivers ; 16(11): e1900322, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31544357

RESUMO

The synthesis of poly[N,N-bis(3-aminopropyl)glycine] (PAPGly) dendrons Gd-based contrast agents (GdCAs) via an orthogonal protection of the different functional groups and an activation/coupling strategy wherein a specific number of synthetic steps add a generation to the existing dendron has been described. The aim of this protocol is to build up two different generations of dendrons (G-0 or dendron's core, and G-1) with peripheral NH2 groups to conjugate a 1,4,7,10-tetraazacyclododecane-1,4,7-triacetic acid (DO3A) derivative and afterwards to chelate with Gd3+ paramagnetic ions. These complexes, which have a well-defined molecular weight, are of relevance to MRI as an attempt to gain higher 1 H relaxivity by slowing down the rotation of molecule compared to monomeric Gd(III) complexes used as contrast agents and to increase the number of paramagnetic centers present in one molecular structure. From the study of their water 1 H longitudinal relaxation rate at different magnetic fields (NMRD, Nuclear Magnetic Relaxation Dispersion) and by evaluating the variable temperature 17 O-NMR data we determined the parameters characterizing the water exchange rate and the rotational correlation time of each complex, both affecting 1 H relaxivity. Furthermore, these two novel PAPGly GdCAs were objects of i) an in vivo study to determine their biodistributions in healthy C57 mice at several time points, and ii) the Dynamic Contrast-Enhanced MRI (DCE-MRI) approach to assess their contrast efficiency measured in the tumor region of C57BL/6 mice transplanted subcutaneously with B16-F10 melanoma cells. The aim of the comparison of these two dendrons GdCAs, having different molecular weights (MW), is to understand how MW and relaxivity may influence the contrast enhancement capabilities in vivo at low magnetic field (1 T). Significant contrast enhancement was observed in several organs (vessel, spleen and liver), already at 5 min post-injection, for the investigated CAs. Moreover, these CAs induced a marked contrast enhancement in the tumor region, thanks to the enhanced permeability retention effect of those macromolecular structures.


Assuntos
Meios de Contraste/química , Gadolínio/química , Melanoma/química , Compostos Organometálicos/química , Animais , Meios de Contraste/síntese química , Meios de Contraste/farmacocinética , Gadolínio/farmacocinética , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Melanoma/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Compostos Organometálicos/síntese química , Compostos Organometálicos/farmacocinética , Distribuição Tecidual
10.
Contrast Media Mol Imaging ; 2019: 5080267, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31182936

RESUMO

The optical imaging plays an increasing role in preclinical studies, particularly in cancer biology. The combined ultrasound and optical imaging, named photoacoustic imaging (PAI), is an emerging hybrid technique for real-time molecular imaging in preclinical research and recently expanding into clinical setting. PAI can be performed using endogenous contrast, particularly from oxygenated and deoxygenated hemoglobin and melanin, or exogenous contrast agents, sometimes targeted for specific biomarkers, providing comprehensive morphofunctional and molecular information on tumor microenvironment. Overall, PAI has revealed notable opportunities to improve knowledge on tumor pathophysiology and on the biological mechanisms underlying therapy. The aim of this review is to introduce the principles of PAI and to provide a brief overview of current PAI applications in preclinical research, highlighting also on recent advances in clinical translation for cancer diagnosis, staging, and therapy.


Assuntos
Neoplasias/diagnóstico , Técnicas Fotoacústicas/métodos , Nanomedicina Teranóstica/tendências , Absorção de Radiação , Animais , Biomarcadores Tumorais/efeitos da radiação , Sistemas Computacionais , Meios de Contraste/efeitos da radiação , Detecção Precoce de Câncer/métodos , Desenho de Equipamento , Hemoglobinas/efeitos da radiação , Humanos , Verde de Indocianina/efeitos da radiação , Lasers , Substâncias Macromoleculares/efeitos da radiação , Melaninas/efeitos da radiação , Nanopartículas Metálicas/efeitos da radiação , Nanotubos de Carbono/efeitos da radiação , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Técnicas Fotoacústicas/instrumentação , Pontos Quânticos/efeitos da radiação , Espalhamento de Radiação , Nanomedicina Teranóstica/métodos , Transdutores
11.
J Am Chem Soc ; 141(21): 8462-8472, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-30925046

RESUMO

Biomedical investigations reveal that excessive formaldehyde generation is possibly a critical factor for tissue cancerization, cancer progression, and metastasis. Responsive molecular probes that can detect lysosomal formaldehyde in live cells and tumors and monitor drug-triggered formaldehyde scavenging contribute potentially to future cancer diagnosis and treatment monitoring. Herein, a novel "dual-key-and-lock" strategy-based ruthenium(II) complex probe, Ru-FA, is reported as an effective tool for formaldehyde detection in vitro and in vivo. Ru-FA shows weak luminescence due to photon-induced electron transfer (PET) process from Ru(II) center to electron withdrawing group 2,4-dinitrobenzene (DNB). Triggered by the specific reaction with formaldehyde (first "key") in an acidic microenvironment (second "key"), DNB is cleaved from Ru-FA, affording an emissive Ru(II) complex derivative, Ru-NR. Spectrometric analysis including steady-state and time-gated luminescence indicates that Ru-FA is favorable to be used as the probe for quantification of formaldehyde in human sera and mouse organs. Ru-FA is biocompatible and cell membrane permeable. Together with its smart "dual-key-and-lock" response to formaldehyde, luminescence imaging of lysosomal formaldehyde in live cells, visualization of tumor-derived endogenous formaldehyde, and monitoring of formaldehyde scavenging in mice were achieved, followed by the successful demonstration on detection of formaldehyde in tumors and other organs. These in vivo and in vitro detection confirm not only the excessive formaldehyde generation in tumors, but also the efficient drug administration to scavenge formaldehyde, demonstrating the potential application of Ru-FA in cancer diagnosis and treatment monitoring through lysosomal formaldehyde detection.


Assuntos
Complexos de Coordenação/química , Formaldeído/análise , Sondas Moleculares/química , Imagem Óptica , Rutênio/química , Neoplasias do Colo do Útero/química , Animais , Complexos de Coordenação/síntese química , Transporte de Elétrons , Feminino , Células HeLa , Humanos , Lisossomos/química , Camundongos , Camundongos Nus , Sondas Moleculares/síntese química , Estrutura Molecular , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias do Colo do Útero/diagnóstico por imagem
12.
Langmuir ; 35(11): 3992-3998, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30844286

RESUMO

Nanoparticles (NPs) of protein-based materials have become one of the most promising candidates for drug carriers in drug-delivery systems because of their in vivo nontoxicity, biodegradability, compatibility with hydrophilic drugs, and adaptability to the human body. Many studies have investigated the fabrication of protein NPs from human serum albumin (HSA) as a new drug carrier. It is important for these NPs to remain in the blood until they reach their therapeutic target to achieve the desired effect; the quicker the clearance of drugs from the body, the shorter is the residence time of drugs in the body, which eventually reduces drug efficacy. Macrophage uptake is a major mechanism for clearance of NPs from the body, so, reducing the degree of macrophage uptake is a major challenge in drug-delivery systems. Original studies of HSA NP uptake by macrophages showed that denatured HSA and HSA NPs synthesized with 80% (v/v) ethanol showed a high degree of macrophage uptake. We found that HSA NPs synthesized with lower ethanol content at pH 7 showed lower macrophage uptake in in vitro macrophage cellular uptake experiments. The effects of the preparation parameters of ethanol concentration, pH, and glutaraldehyde on the macrophage uptake of NPs were thoroughly studied. This newly developed protein NP with lower macrophage uptake has potential application as a drug carrier for many delivery systems.


Assuntos
Sistema Fagocitário Mononuclear , Nanopartículas/química , Albumina Sérica Humana/química , Animais , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos , Etanol/química , Feminino , Glutaral/química , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Macrófagos/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Células PC-3 , Tamanho da Partícula , Albumina Sérica Humana/síntese química , Albumina Sérica Humana/metabolismo , Propriedades de Superfície , Distribuição Tecidual
13.
Biochem Biophys Res Commun ; 508(2): 646-653, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30527804

RESUMO

Armadillo-related proteins function in both signal transduction and cell adhesion, it also plays a central role in tumorigenesis. Plakophilin 3 (PKP3) is a member of the armadillo protein family. PKP3 has demonstrated a role in melanoma, breast cancer, gastric cancer, and other kind of cancers; however its role in ovarian cancer was not fully understood. In this study we explored the function and mechanisms of PKP3 in ovarian cancer. An elevated level of PKP3 was found in ovarian cancer tissues compared with normal tissues. PKP3 also modulate cellular proliferation and invasion in ovarian cancer. The ability of cellular proliferation, formation, and invasion was significantly decreased after the silencing of PKP3 in SKOV3 cells. While an over-expression of PKP3 in A2780 cells up-regulates the ability of cellular proliferation, formation, and invasion. As for the mechanism of PKP3, mTOR pathway was activated to regulate autophagy according to the interaction of PKP3 with the upstream of MAPK pathway. The result of this study support PKP3 as the oncogene candidate and a potential target for the treatment of ovarian cancer.


Assuntos
Autofagia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Placofilinas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Movimento Celular , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Ovarianas/química , Placofilinas/análise , Placofilinas/genética
14.
J Biotechnol ; 290: 53-58, 2019 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-30579890

RESUMO

We report the design and characterization of novel fusion proteins, consisting of the F8 antibody and of murine calreticulin (Calr). The F8 antibody recognizes the alternatively-spliced ED-A domain of fibronectin, an extracellular matrix component found in most tumor types, while calreticulin has previously been described as an "eat-me" signal for dendritic cells and phagocytes. Four fusion proteins, differing in antibody formats and peptide linkers, were produced in mammalian cells, purified to homogeneity and tested in vitro and in vivo. A quantitative biodistribution in F9 tumor-bearing mice revealed that the homobivalent F8-F8-Calr format, featuring a tandem diabody structure, had the best tumor-homing properties and, for this reason, this protein was studied in therapy experiments in CT26 tumor-bearing mice. Intravenous administration of F8-F8-Calr led to a tumor growth retardation, which could be further improved by combination with anti-PD1 antibody treatment. Immunohistochemical analysis revealed an increased density of CD8+ T cells, CD11c+ dendritic cells and F4/80+ macrophages in tumor tissue. Even though F8-F8-Calr did not lead to cancer cures at the doses tested, the excellent tolerability profile and the ability to favor a leukocyte infiltration into the neoplastic mass suggests that the targeted delivery of calreticulin may be considered for combination therapy approaches.


Assuntos
Antineoplásicos/metabolismo , Calreticulina/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Fibronectinas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Antineoplásicos/farmacocinética , Células CHO , Calreticulina/química , Calreticulina/genética , Cricetinae , Cricetulus , Feminino , Fibronectinas/química , Fibronectinas/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual
15.
J Surg Oncol ; 118(2): 253-264, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30080930

RESUMO

Fluorescence-guided surgery can enhance the surgeon's ability to achieve a complete oncologic resection. There are a number of tumor-specific probes being developed with many preclinical mouse models to evaluate their efficacy. The current review discusses the different preclinical mouse models in the setting of probe evaluation and highlights the advantages of patient-derived orthotopic xenografts (PDOX) mouse models and genetic reporters to develop fluorescence-guided surgery.


Assuntos
Proteínas Luminescentes/análise , Neoplasias Experimentais/química , Neoplasias Experimentais/cirurgia , Imagem Óptica/métodos , Cirurgia Assistida por Computador/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Linhagem Celular Tumoral , Genes Reporter , Engenharia Genética/métodos , Xenoenxertos/patologia , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias/métodos , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo
16.
Anal Chem ; 90(10): 6051-6058, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29668267

RESUMO

Described is a quantitative-mass-spectrometry-imaging (qMSI) methodology for the analysis of lactate and glutamate distributions in order to delineate heterogeneity among mouse tumor models used to support drug-discovery efficacy testing. We evaluate and report on preanalysis-stabilization methods aimed at improving the reproducibility and efficiency of quantitative assessments of endogenous molecules in tissues. Stability experiments demonstrate that optimum stabilization protocols consist of frozen-tissue embedding, post-tissue-sectioning desiccation, and storage at -80 °C of tissue sections sealed in vacuum-tight containers. Optimized stabilization protocols are used in combination with qMSI methodology for the absolute quantitation of lactate and glutamate in tumors, incorporating the use of two different stable-isotope-labeled versions of each analyte and spectral-clustering performed on each tissue section using k-means clustering to allow region-specific, pixel-by-pixel quantitation. Region-specific qMSI was used to screen different tumor models and identify a phenotype that has low lactate heterogeneity, which will enable accurate measurements of lactate modulation in future drug-discovery studies. We conclude that using optimized qMSI protocols, it is possible to quantify endogenous metabolites within tumors, and region-specific quantitation can provide valuable insight into tissue heterogeneity and the tumor microenvironment.


Assuntos
Ácido Glutâmico/análise , Ácido Láctico/análise , Espectrometria de Massas , Animais , Feminino , Ácido Glutâmico/metabolismo , Ácido Láctico/metabolismo , Camundongos , Camundongos Nus , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo
17.
Anal Chem ; 90(4): 2648-2654, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29359562

RESUMO

The development of novel fluorescent probes for monitoring the concentration of various biomolecules in living systems has great potential for eventual early diagnosis and disease intervention. Selective detection of competitive species in biological systems is a great challenge for the design and development of fluorescent probes. To improve on the design of fluorescent coumarin-based biothiol sensing technologies, we have developed herein an enhanced dual emission doubly activated system (DACP-1 and the closely related DACP-2) for the selective detection of glutathione (GSH) through the use of one optical channel and the detection of cysteine (Cys) by another channel. A phenylselenium group present at the 4-position completely quenches the fluorescence of the probe via photoinduced electron transfer to give a nonfluorescent species. Probes are selective for glutathione (GSH) in the red region and for cysteine/homocysteine (Cys/Hcy) in the green region. When they were treated with GSH, DACP-1 and DACP-2 showed strong fluorescence enhancement in comparison to that for closely related species such as amino acids, including Cys/Hcy. Fluorescence quantum yields (ΦF) increased for the red channel (<0.001 to 0.52 (DACP-1) and 0.48 (DACP-2)) and green channel (Cys) (<0.001 to 0.030 (DACP-1) and 0.026 (DACP-2)), respectively. Competing fluorescent enhancements upon addition of closely related species were negligible. Fast responses, improved water solubility, and good cell membrane permeability were all properly established with the use of DACP-1 and DACP-2. Live human lung cancer cells and fibroblasts imaged by confocal microscopy, as well as live mice tumor model imaging, confirmed selective detection.


Assuntos
Cisteína/análise , Fibroblastos/química , Corantes Fluorescentes/química , Glutationa/análise , Neoplasias Pulmonares/química , Imagem Óptica , Animais , Sobrevivência Celular/efeitos dos fármacos , Teoria da Densidade Funcional , Feminino , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/farmacologia , Humanos , Injeções Intravenosas , Neoplasias Pulmonares/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Confocal , Estrutura Molecular , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Células Tumorais Cultivadas
18.
IEEE Trans Med Imaging ; 37(1): 241-250, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29293430

RESUMO

Ultrasound molecular imaging (USMI) is accomplished by detecting microbubble (MB) contrast agents that have bound to specific biomarkers, and can be used for a variety of imaging applications, such as the early detection of cancer. USMI has been widely utilized in preclinical imaging in mice; however, USMI in humans can be challenging because of the low concentration of bound MBs and the signal degradation caused by the presence of heterogenous soft tissue between the transducer and the lesion. Short-lag spatial coherence (SLSC) beamforming has been proposed as a robust technique that is less affected by poor signal quality than standard delay-and-sum (DAS) beamforming. In this paper, USMI performance was assessed using contrast-enhanced ultrasound imaging combined with DAS (conventional CEUS) and with SLSC (SLSC-CEUS). Each method was characterized by flow channel phantom experiments. In a USMI-mimicking phantom, SLSC-CEUS was found to be more robust to high levels of additive thermal noise than DAS, with a 6dB SNR improvement when the thermal noise level was +6dB or higher. However, SLSC-CEUS was also found to be insensitive to increases in MB concentration, making it a poor choice for perfusion imaging. USMI performance was also measured in vivo using VEGFR2-targeted MBs in mice with subcutaneous human hepatocellular carcinoma tumors, with clinical imaging conditions mimicked using a porcine tissue layer between the tumor and the transducer. SLSC-CEUS improved the SNR in each of ten tumors by an average of 41%, corresponding to 3.0dB SNR. These results indicate that the SLSC beamformer is well-suited for USMI applications because of its high sensitivity and robust properties under challenging imaging conditions.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Modelos Biológicos , Imagem Molecular/métodos , Ultrassonografia/métodos , Animais , Artefatos , Xenoenxertos/química , Xenoenxertos/diagnóstico por imagem , Humanos , Camundongos , Neoplasias Experimentais/química , Neoplasias Experimentais/diagnóstico por imagem , Imagens de Fantasmas , Sensibilidade e Especificidade , Razão Sinal-Ruído , Suínos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Adv Healthc Mater ; 6(15)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28544639

RESUMO

The "Tumor microenvironment" (TME) is a complex, interacting system of the tumor and its surrounding environment. The TME has drawn more attention recently in attempts to overcome current drug resistance and the recurrence of cancer by understanding the cancer and its microenvironment systematically, beyond past reductionist approaches. However, a lack of experimental tools to dissect the intricate interactions has hampered in-depth research into the TME. Here, a biomimetic TME model using a microfluidic platform is presented, which enables the interaction between TME constituents to be studied in a comprehensive manner. Paracrine interactions of cocultured tumor cell lines (SK-OV-3, MKN-74, and SW620) with primary fibroblasts show marked morphological changes in the tumor cells, depending on the type of tumor cells, and, importantly, the composition of the extracellular matrix. Furthermore, this model allows direct observation of angiogenesis induced by the tumor-stroma interaction. Finally, reconstituting simultaneous angiogenesis and lymphangiogenesis induced by the tumor-stromal interaction with TME mimicking extrinsic factors is enabled. It is believed that the in vitro biomimetic model and the experimental concepts described will help to shed light on the complex biology of the TME.


Assuntos
Técnicas de Cultura Celular por Lotes/instrumentação , Materiais Biomiméticos/química , Dispositivos Lab-On-A-Chip , Neoplasias Experimentais/química , Neoplasias Experimentais/fisiopatologia , Engenharia Tecidual/instrumentação , Microambiente Tumoral , Técnicas de Cultura Celular por Lotes/métodos , Linhagem Celular Tumoral , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Teste de Materiais , Neoplasias Experimentais/patologia , Engenharia Tecidual/métodos
20.
Angew Chem Int Ed Engl ; 56(15): 4314-4319, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28295960

RESUMO

The success of nanomedicines in the clinic depends on our comprehensive understanding of nano-bio interactions in tumor microenvironments, which are characterized by dense leaky microvasculature and acidic extracellular pH (pHe ) values. Herein, we investigated the accumulation of ultrasmall renal-clearable gold NPs (AuNPs) with and without acidity targeting in xenograft mouse models of two prostate cancer types, PC-3 and LNCaP, with distinct microenvironments. Our results show that both sets of AuNPs could easily penetrate into the tumors but their uptake and retention were mainly dictated by the tumor microvasculature and the enhanced permeability and retention effect over the entire targeting process. On the other hand, increased tumor acidity indeed enhanced the uptake of AuNPs with acidity targeting, but only for a limited period of time. By making use of simple surface chemistry, these two effects can be synchronized in time for high tumor targeting, opening new possibilities to further improve the targeting efficiencies of nanomedicines.


Assuntos
Ouro/farmacocinética , Rim/metabolismo , Nanopartículas Metálicas/química , Neoplasias da Próstata/química , Microambiente Tumoral , Animais , Ouro/química , Ouro/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Rim/química , Masculino , Camundongos , Nanomedicina , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Neoplasias da Próstata/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA