Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.731
Filtrar
1.
Bull Exp Biol Med ; 175(2): 254-259, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37466854

RESUMO

We studied the effects of the extract of the terrestrial part of Aconitum baicalense in BALB/c female mice at the early stages after the injection of N-methyl-N-nitrosourea (MNU). The extract reduced inflammatory activity and tumor growth in the mammary gland. The antitumor and anti-inflammatory effects of the extract are based on the inhibition of cancer stem cells, hematopoietic stem cells, and hematopoietic progenitor cells that promote inflammation. The extract of A. baicalense disrupted the recruitment of epithelial progenitor cells and angiogenesis precursors to the mammary gland preventing neovascularization and transformation of epithelial cells into tumor cells.


Assuntos
Aconitum , Células-Tronco Adultas , Neoplasias Mamárias Experimentais , Feminino , Camundongos , Animais , Metilnitrosoureia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Células-Tronco Adultas/patologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia
2.
Sci Rep ; 13(1): 7703, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37169856

RESUMO

Breast cancer is the second leading cause of cancer death among women. The present study is an effort to reveal the antiproliferative and antioxidant actions of mango seed kernel extract (KE), peel extract (PE), and their combination (KEPE) on mammary tumors induced by 7,12 dimethylbenz[a]anthracene (DMBA). Seven groups of adult female Sprague-Dawley rats were prepared, including C: (control), DMBA: (rats were administered with DMBA), (DMBA-KE), (DMBA-PE), and (DMBA-KEPE): rats were administered with DMBA and then treated with KE, PE, and (both KE and PE), respectively, (KE) and (PE): rats were administered with KE and PE, separately. The study focused on the assessment of markers of endocrine derangement [serum 17-ß estradiol (E2)], apoptosis [caspase-3 and deoxyribonucleic acid fragmentation (DNAF)], and oxidative stress [lipid peroxidation and antioxidants (glutathione, glutathione-S-transferase, glutathione reductase, glutathione peroxidase, and superoxide dismutase)]. Histopathological examination and immunohistochemical expression of caspase-3 and estrogen receptor-α (ER-α) in mammary gland tissues (MGTs) were determined, as well as the characterization of mango extracts. The results showed that DMBA administration induced mammary tumors by increasing cell proliferation and evading apoptosis. In addition, DMBA administration caused oxidative stress by the production of reactive oxygen species, which increased lipid peroxidation and decreased cellular antioxidants, allowing cancer to progress. In contrast, treatment with DMBA-KE, DMBA-PE, or DMBA-KEPE diminished mammary tumors induced by DMBA, where they reduced oxidative stress via increased antioxidant parameters including reduced glutathione, superoxide dismutase, total glutathione peroxidase, glutathione reductase, and glutathione S-transferase. Also, different treatments decreased proliferation through the reduction of E2, and ER-α expression levels. However, these treatments increased the apoptosis of unwanted cells as they increased caspase-3 activity and DNAF. All these changes led to the prevention of breast injuries and the reduction of mammary tumors. This demonstrates that the contents of mango extracts, especially phenolics and flavonoids, have an important role in mammary tumor treatment through their potential antioxidant, antiproliferative, proapoptotic, and anti-estrogenic effects. KE and PE administration for 4 weeks had no adverse effects. Conclusion: Each of KE, PE, and KEPE has a therapeutic effect against DMBA-induced mammary tumors via induction of apoptosis and reduction of each of the OS, proliferation, and estrogenic effects. So, they can play an important role in the pharmacological tole.


Assuntos
Neoplasias Mamárias Experimentais , Mangifera , Ratos , Feminino , Animais , Antioxidantes/metabolismo , Ratos Sprague-Dawley , Mangifera/metabolismo , Caspase 3 , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/prevenção & controle , Glutationa , Superóxido Dismutase , Carcinogênese , Oxirredutases
3.
Nutr Cancer ; 75(7): 1579-1599, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37255368

RESUMO

Among luminal types of breast cancers, ER + breast cancer is the most frequently diagnosed cancer globally. ER + breast cancer is commonly treated with SERM drugs that block ER to prevent ER-mediated cancerous growth. Our previous computational screening found pelargonidin (PG) can inhibit ER-signaling with potent bioactivity and satisfactory toxicological features. The present study explored the anti-tumoral prospect of PG against DMBA-induced ER + murine mammary carcinogenesis. The female BALB/c mice were divided into control (A) and DMBA-exposed groups. Following tumor appearance, the DMBA-exposed group was divided into five groups: tumor control, PG-treated (Groups P25, P50, and P100), and tamoxifen-treated (TAM). The results indicated that PG-treatment dose-dependently reduced the mean tumor volume, reinstated body weight loss, and enhanced the percentage survival of tumor-bearing mice. In addition, we recorded a significant reduction in LPO, total cholesterol, and triglycerides and a surge in the activity of antioxidases and phase II detoxifying enzymes in PG-treated animals. PG also dose-dependently increased the serum level of unbound estradiol, an indicator of competitive ER binding by an ER agonist/antagonist. These data suggest that pelargonidin has potent anticancer potential against the animal model of ER + breast cancer that matches the efficiency of tamoxifen with conceivably fewer side effects.


Assuntos
Neoplasias Mamárias Experimentais , Tamoxifeno , Camundongos , Feminino , Animais , Camundongos Endogâmicos BALB C , Tamoxifeno/farmacologia , Transformação Celular Neoplásica , Estresse Oxidativo , Lipídeos/farmacologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/prevenção & controle , 9,10-Dimetil-1,2-benzantraceno/toxicidade
4.
Int J Mol Sci ; 24(6)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36982737

RESUMO

Estrogen receptor-positive breast cancers (ER+ BCas) are the most common form of BCa and are increasing in incidence, largely due to changes in reproductive practices in recent decades. Tamoxifen is prescribed as a component of standard-of-care endocrine therapy for the treatment and prevention of ER+ BCa. However, it is poorly tolerated, leading to low uptake of the drug in the preventative setting. Alternative therapies and preventatives for ER+ BCa are needed but development is hampered due to a paucity of syngeneic ER+ preclinical mouse models that allow pre-clinical experimentation in immunocompetent mice. Two ER-positive models, J110 and SSM3, have been reported in addition to other tumour models occasionally shown to express ER (for example 4T1.2, 67NR, EO771, D2.0R and D2A1). Here, we have assessed ER expression and protein levels in seven mouse mammary tumour cell lines and their corresponding tumours, in addition to their cellular composition, tamoxifen sensitivity and molecular phenotype. By immunohistochemical assessment, SSM3 and, to a lesser extent, 67NR cells are ER+. Using flow cytometry and transcript expression we show that SSM3 cells are luminal in nature, whilst D2.0R and J110 cells are stromal/basal. The remainder are also stromal/basal in nature; displaying a stromal or basal Epcam/CD49f FACS phenotype and stromal and basal gene expression signatures are overrepresented in their transcript profile. Consistent with a luminal identity for SSM3 cells, they also show sensitivity to tamoxifen in vitro and in vivo. In conclusion, the data indicate that the SSM3 syngeneic cell line is the only definitively ER+ mouse mammary tumour cell line widely available for pre-clinical research.


Assuntos
Neoplasias da Mama , Receptores de Estrogênio , Tamoxifeno , Humanos , Linhagem Celular Tumoral , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Animais , Camundongos , Modelos Animais de Doenças , Receptores de Estrogênio/genética , Tamoxifeno/farmacologia , Fenótipo , Imuno-Histoquímica , Citometria de Fluxo , Transcriptoma , Camundongos da Linhagem 129 , RNA-Seq , Células Epiteliais , Glândulas Mamárias Animais/citologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética
5.
Biomed Pharmacother ; 155: 113675, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36115110

RESUMO

BACKGROUND: Breast cancer prevalence has been globally increasing, therefore, introducing novel interventions in cancer treatment is of a significant importance. The present study was designed to investigate the anti-cancer effect of Canagliflozin (CNG) in an experimental model of DMBA-induced mammary carcinoma in female rats. METHODS: 18 female rats were divided into three experimental groups: Normal control, DMBA control, DMBA+ CNG treated group. DMBA (7.5 mg/kg) was injected subcutaneously in the mammary cells twice weekly for 4 weeks and CNG (10 mg/kg) was orally administered daily for an additional 3 weeks while DMBA control rats only received the vehicle for 3 weeks. Tumors' weight and volume were measured, BRCA-1 and TAC were quantified in serum samples, mTOR, caspase-1, NFκB, IL-1ß, NLRP3, GSDMD and MDA were quantified in tumors' homogenates. RESULTS: CNG treatment increased the BRCA-1 expression, suppressed mTOR inflammatory pathway, attenuated tumor inflammatory mediators; NLRP3, GSDMD, NFκB, IL-1ß, suppressed the oxidative stress and inhibited tumor expression of the proliferation biomarker; Ki67. CONCLUSION: CNG modulated mTOR-mediated signaling pathway and attenuated pyroptotic, inflammatory pathways, suppressed oxidative stress and eventually inhibited DMBA-induced mammary carcinoma proliferation.


Assuntos
Carcinoma , Neoplasias Mamárias Experimentais , Ratos , Feminino , Animais , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Antígeno Ki-67/metabolismo , Canagliflozina , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/prevenção & controle , Ratos Sprague-Dawley , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais , Caspase 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mediadores da Inflamação
6.
Pak J Biol Sci ; 25(6): 485-494, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36098183

RESUMO

<b>Background and Objective:</b> For more than a decade, breast cancer has been one of the most common forms of cancer among women around the world. The present article aimed to evaluate the protective activity of CEG-AgNPs against DMBA-induced mammary carcinoma. <b>Materials and Methods:</b> In this experimental study, green synthesis and characterization of CEG-AgNPs were carried as well as IC<sub>50</sub> against Mcf7 cell line and LD<sub>50</sub> on mice were evaluated. A total of 24 adult albino mice were divided into four groups six rats in each. Group I was given an equal amount of distilled water, group II was received 80 mg kg<sup></sup><sup>1</sup> b.wt., DMBA for 4 weeks, groups III and IV were treated with CEG-AgNPs (28.1 and 70.25 mg kg<sup></sup><sup>1</sup>) from the 5th week of DMBA administration for 4 weeks, respectively. <b>Results:</b> CEG-AgNPs were approximately 42.32±9.52 nm with a negative zeta potential of -17.44. It is IC<sub>50</sub> against the Mcf7 cell line and LD<sub>50</sub> is equal to 82.76 µg mL<sup></sup><sup>1</sup> and 1405 mg kg<sup></sup><sup>1</sup> b.wt., A significant normalization in plasma ALT, AST, AST and LDH as well as mammary MDA, TNF-α, IL-6, P53, SOD, GPx and GSH levels have been observed in CEG-AgNPs treated mice. Oral CEG-AgNPs administration has suppressed VEGF-C gene expression in DMBA-treated mice. <b>Conclusion:</b> The present results, biochemical, histological and MRI results showed that CEG-AgNPs have potent anticancer activity against DMBA-induced mammary carcinoma in mice by inducing the biosynthesizes of antioxidant biomarkers and suppression of cytokines gene expression.


Assuntos
Carcinoma , Neoplasias Mamárias Experimentais , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Antioxidantes , Citocinas , Feminino , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Ratos
7.
J Biochem Mol Toxicol ; 36(10): e23146, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35698847

RESUMO

Breast cancer is the primary cause of cancer-related death in females, wherein increased mortality of breast cancer patients is recorded worldwide. Zingiberene is a monocyclic sesquiterpene from the ginger plant and has many pharmacological benefits. In this exploration, we assessed the anticancer actions of Zingiberene against the 7,12-dimethylbenz(a)anthracene (DMBA)-stimulated mammary carcinogenesis in rats and MDA-MB-231 cells. Breast cancer was induced in the Female Sprague-Dawley rats through the 25 mg/kg of DMBA in 0.5 ml of corn oil and then treated with 20 and 40 mg/kg of Zingiberene, respectively. The body weight of animals and tumor volume was measured. Hematological parameters, transaminases, lipid profile, lipid peroxidation, and antioxidants status were scrutinized using standard techniques. The estrogen receptor-α and inflammatory markers were inspected by using respective assay kits. Histological damage scores were determined. In vitro experiments were conducted to scrutinize Zingiberene's effect on cell viability and apoptotic cell death in MDA-MB-231 cells. Zingiberene substantially modulated the DMBA-stimulated physiological and hematological changes and decreased the transaminases, and lipid peroxidation in the DMBA-stimulated animals. Zingiberene also elevated the antioxidant level and suppressed the inflammatory markers. Histological study revealed the protective effects of Zingiberene. The viability of MDA-MB-231 cells was noticeably diminished by the Zingiberene, thus inducing apoptotic cell death. Overall, our findings reliably proved the anticancer potential of Zingiberene against the DMBA-stimulated mammary tumorigenesis, and it could be a promising chemotherapeutic agent.


Assuntos
9,10-Dimetil-1,2-benzantraceno , Neoplasias Mamárias Experimentais , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Antracenos , Antioxidantes/metabolismo , Carcinógenos , Óleo de Milho/efeitos adversos , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/prevenção & controle , Sesquiterpenos Monocíclicos , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio , Transaminases
8.
Breast Cancer Res ; 24(1): 41, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715861

RESUMO

BACKGROUND: The majority of BRCA1-mutant breast cancers are characterized by a triple-negative phenotype and a basal-like molecular subtype, associated with aggressive clinical behavior. Current treatment options are limited, highlighting the need for the development of novel targeted therapies for this tumor subtype. METHODS: Our group previously showed that EZH2 is functionally relevant in BRCA1-deficient breast tumors and blocking EZH2 enzymatic activity could be a potent treatment strategy. To validate the role of EZH2 as a therapeutic target and to identify new synergistic drug combinations, we performed a high-throughput drug combination screen in various cell lines derived from BRCA1-deficient and -proficient mouse mammary tumors. RESULTS: We identified the combined inhibition of EZH2 and the proximal DNA damage response kinase ATM as a novel synthetic lethality-based therapy for the treatment of BRCA1-deficient breast tumors. We show that the combined treatment with the EZH2 inhibitor GSK126 and the ATM inhibitor AZD1390 led to reduced colony formation, increased genotoxic stress, and apoptosis-mediated cell death in BRCA1-deficient mammary tumor cells in vitro. These findings were corroborated by in vivo experiments showing that simultaneous inhibition of EZH2 and ATM significantly increased anti-tumor activity in mice bearing BRCA1-deficient mammary tumors. CONCLUSION: Taken together, we identified a synthetic lethal interaction between EZH2 and ATM and propose this synergistic interaction as a novel molecular combination for the treatment of BRCA1-mutant breast cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas Mutadas de Ataxia Telangiectasia , Proteína BRCA1 , Neoplasias da Mama , Proteína Potenciadora do Homólogo 2 de Zeste , Indóis , Inibidores de Proteínas Quinases , Piridonas , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA1/deficiência , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Humanos , Indóis/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Piridonas/farmacologia , Mutações Sintéticas Letais
9.
Cancer Immunol Res ; 10(6): 672-673, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35544128

RESUMO

Immune checkpoint inhibitors have yet to significantly improve outcomes for hormone-dependent estrogen/progesterone receptor-positive breast cancer. To address this issue, there is a need for murine models that more closely mimic hormone receptor-positive breast cancer. In this issue, Gil Del Alcazar and colleagues provide an in-depth characterization of a Nitroso-N-methylurea-induced mammary tumor model in outbred Sprague-Dawley rats that meets these needs as it mimics the heterogeneity for mutational profiles, estrogen receptor expression, and immune evasive mechanisms observed in human breast cancer. See related article by Gil Del Alcazar et al., p. 680 (1).


Assuntos
Neoplasias da Mama , Neoplasias Mamárias Experimentais , Animais , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Imunoterapia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Progesterona/efeitos adversos , Ratos , Ratos Sprague-Dawley
10.
Nanoscale ; 14(17): 6312-6322, 2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35393985

RESUMO

Objectives: To determine whether two-step ablation using sequential low and high temperature heating can achieve improved outcomes in animal tumor models when combined with chemotherapeutic liposomes (LP). Materials and methods: Balb/c mice bearing 4T1 tumor received paclitaxel-loaded liposomes followed 24 h later by either traditional RFA (70 °C, 5 min) or a low temperature RFA (45 °C, 5 min), or two-step RFA (45 °C 2 min + 70 °C 3 min). Intratumoral drug accumulation and bio-distribution in major organs were evaluated. Periablational drug penetration was evaluated by pathologic staining and the intratumoral interstitial fluid pressure (IFP) was measured directly. For long-term outcomes, mice bearing 4T1 or H22 tumors were randomized into five groups (n = 8 per group): control (no treatment), RFA alone, LP + RFA (45 °C), LP + RFA (70 °C) and LP + RFA (45 + 70 °C). End-point survivals were compared among the different groups. Results: The greater intratumoral drug accumulation (3.35 ± 0.32 vs. 3.79 ± 0.29 × 108 phot/cm2/s at 24 h, p = 0.09), deeper periablational drug penetration (45.7 ± 5.0 vs. 1.6 ± 0.5, p < 0.001), and reduced off-target drug deposition in major organs (liver 96.1 ± 31.6 vs. 47.4 ± 1.5 × 106 phot/cm2/s, p < 0.001) were found when combined with RFA (45 °C) compared to drug alone. For long-term outcomes, 4T1 tumor growth rates for LP + two-step RFA (45 + 70 °C) were significantly slower than those of LP + RFA (70 °C), LP + RFA (45 °C), and RFA alone (P < 0.01 for all comparisons). End point survival for LP + RFA (45 + 70 °C) was also longer than that for LP + RFA (70 °C) (median 16 vs. 10 days, p = 0.003) or LP + RFA 45 °C (11 days, p = 0.009) and RFA alone (8.3 days, p < 0.001) in 4T1 tumor models. The intratumoral IFP after RFA (45 °C) was significantly lower than baseline RFA (3.3 ± 0.8 vs. 19.2 ± 3.1 mmHg, p < 0.001), but was not measurable after RFA (70 °C). Conclusions: A two-step ablation combined with chemotherapeutic liposomes can achieve better survival benefit compared to traditional RFA in animal models.


Assuntos
Ablação por Cateter , Neoplasias Mamárias Experimentais , Animais , Lipossomos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico
11.
Biomed Pharmacother ; 147: 112666, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35124384

RESUMO

Naturally occurring phytochemicals especially polyphenolic compounds have received increasing attention as chemopreventive agents. The chemopreventive potential of the ethanolic extract of Salvadora persica L. fruits SP, (the arak tree or miswak) on 7,12-dimethylbenz (a) anthracene (DMBA)-induced mammary carcinogenesis in female albino rats was investigated in this work. Ethanolic extract of SP fruits was supplemented to the experimental groups at a concentration of 500 mg/kg body weight for 22 weeks. Administration of SP extract suppressed DMBA-induced mammary carcinogenesis as revealed by incidence of tumors in histological investigation. There was a significant reduction in cell proliferation and an increase in apoptosis with downregulation of estrogen receptor expression in the mammary tissue of SP-treated animals. Additionally, SP extract prevented the oxidative damage induced in breast tissues of DMBA-treated rats. SP treatment also decreased the viability of MCF-7 breast cancer cells and induced early and late apoptosis and induced S cell cycle arrest. The chemo-preventive properties and anticancer effects of SP could be attributed to its anti-oxidative and a high percentage of phenolic compounds and esters which were detected here in the SP fruit extract.


Assuntos
Neoplasias Mamárias Experimentais/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Salvadoraceae , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Distribuição Aleatória , Ratos , Ratos Wistar , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Molecules ; 27(3)2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35164019

RESUMO

Breast cancer is a major cause of death in women worldwide. In this study, 60 female rats were classified into 6 groups; negative control, α-aminophosphonates, arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one, DMBA, DMBA & α-aminophosphonates, and DMBA & arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. New α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one were synthesized and elucidated by different spectroscopic and elemental analysis. Histopathological examination showed marked proliferation of cancer cells in the DMBA group. Treatment with α-aminophosphonates mainly decreased tumor mass. Bcl2 expression increased in DMBA-administered rats and then declined in the treated groups, mostly with α-aminophosphonates. The level of CA15-3 markedly declined in DMBA groups treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. Gene expression of GST-P, PCNA, PDK, and PIK3CA decreased in the DMBA group treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one, whereas PIK3R1 and BAX increased in the DMBA group treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. The molecular docking postulated that the investigated compounds can inhibt the Thymidylate synthase TM due to high hydrophobicity charachter.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Timidilato Sintase/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Antineoplásicos/farmacologia , Células CACO-2 , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Feminino , Peixes , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Terapia de Alvo Molecular/métodos , Organofosfonatos/síntese química , Organofosfonatos/química , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Extratos Vegetais , Quinolinas/síntese química , Quinolinas/química , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Ratos , Timidilato Sintase/química
13.
Cell Mol Life Sci ; 79(1): 34, 2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-34989869

RESUMO

New therapeutic targets that could improve current antitumor therapy and overcome cancer resistance are urgently needed. Promising candidates are lysosomal cysteine cathepsins, proteolytical enzymes involved in various critical steps during cancer progression. Among them, cathepsin X, which acts solely as a carboxypeptidase, has received much attention. Our results indicate that the triazole-based selective reversible inhibitor of cathepsin X named Z9 (1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-((4-isopropyl-4H-1,2,4-triazol-3-yl)thio)ethan-1-one) significantly reduces tumor progression, both in vitro in cell-based functional assays and in vivo in two independent tumor mouse models: the FVB/PyMT transgenic and MMTV-PyMT orthotopic breast cancer mouse models. One of the mechanisms by which cathepsin X contributes to cancer progression is the compensation of cathepsin-B activity loss. Our results confirm that cathepsin-B inhibition is compensated by an increase in cathepsin X activity and protein levels. Furthermore, the simultaneous inhibition of both cathepsins B and X with potent, selective, reversible inhibitors exerted a synergistic effect in impairing processes of tumor progression in in vitro cell-based assays of tumor cell migration and spheroid growth. Taken together, our data demonstrate that Z9 impairs tumor progression both in vitro and in vivo and can be used in combination with other peptidase inhibitors as an innovative approach to overcome resistance to antipeptidase therapy.


Assuntos
Catepsina B/antagonistas & inibidores , Catepsinas/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Animais , Catepsina B/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Inibidores Enzimáticos/química , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos Transgênicos , Invasividade Neoplásica , Infiltração de Neutrófilos/efeitos dos fármacos
14.
ACS Appl Mater Interfaces ; 14(1): 417-427, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34978427

RESUMO

The application of drug delivery system (DDS) has achieved breakthroughs in many aspects, especially in the field of tumor treatment. In this work, polyethylene glycol (PEG)-modified hollow mesoporous manganese dioxide (HMnO2@PEG) nanoparticles were used to load the anti-tumor drug bleomycin (BLM). When the DDS reached the tumor site, HMnO2@PEG was degraded and reduced to Mn2+ by the overexpression of glutathione in the tumor microenvironment, and the drug was released simultaneously. BLM coordinated with Mn2+ in situ, thereby greatly improving the therapeutic activity of BLM. The results of in vivo and in vitro treatment experiments showed that the DDS had excellent responsive therapeutic activation ability. In addition, Mn2+ exhibited strong paramagnetism and was used for T1-weighted magnetic resonance imaging in vivo. Furthermore, this therapeutic mode of responsively releasing drugs and activating in situ effectively attenuated pulmonary fibrosis initiated by BLM. In short, this DDS could help in avoiding the side effects of drugs.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Materiais Biocompatíveis/química , Bleomicina/farmacologia , Sistemas de Liberação de Medicamentos , Glutationa/química , Animais , Antibióticos Antineoplásicos/química , Materiais Biocompatíveis/farmacologia , Bleomicina/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Teste de Materiais , Camundongos , Camundongos Nus , Óxidos/química , Óxidos/farmacologia , Tamanho da Partícula , Microambiente Tumoral/efeitos dos fármacos
15.
ACS Appl Mater Interfaces ; 14(3): 3875-3884, 2022 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-35021621

RESUMO

Nanomaterials in combination with radionuclide therapy (RNT) provide new opportunities for cancer treatment. However, nanomaterials with efficient tumor accumulation have been less exploited for effective radionuclide-based therapy. Here, we report glycol chitosan-based nanoparticles (GCP-NPs) with acidic pH-dependent surface charge conversion for efficient radionuclide-based combination therapy. The nanoplatform can change the surface charge of nanoparticles from slight negative to positive in the acidic tumor microenvironment, which facilitates cellular internalization and penetration and thus improves the tumor accumulation efficiency of nanomaterials. Radiolabeling of GCP-NPs with 99mTc enables in vivo radioactive imaging in the mouse subcutaneous tumor model, showing 8.1-fold enhanced tumor uptake relative to pH-insensitive control nanoparticles (termed as GCOP-NPs). Afterward, therapeutic radioisotope 177Lu-labeled GCP-NPs (177Lu-GCP-NPs) that utilize RNT synergistic with photodynamic therapy (PDT) derived from conjugated pyropheophorbide-a within nanoparticles endow superior antitumor efficacy in living cells and tumor-bearing mouse model. More importantly, the combination of RNT and PDT using 177Lu-GCP-NPs can effectively inhibit lung metastasis and eliminate splenomegaly, which is not possible for individual RNT or PDT. Therefore, this study proposes a facile radionuclide-based combination therapy strategy toward complete cancer remission.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Lutécio/química , Nanopartículas/química , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Radioisótopos/química , Animais , Antineoplásicos/química , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Concentração de Íons de Hidrogênio , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Teste de Materiais , Camundongos , Estrutura Molecular , Fármacos Fotossensibilizantes/química
16.
J Mater Chem B ; 10(4): 646-655, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34994759

RESUMO

Nanomedicine-based tumor-targeted therapy has emerged as a promising strategy to overcome the lack of specificity of conventional chemotherapeutic agents. "Passive" targeting caused by the tumor EPR effect and "active" targeting endowed by the tumor-targeting moieties provide promising biomedical utilities and cancer therapy strategies for nanomedicine. However, as the nanoparticles are exposed to biological fluids, a large number of protein molecules will be adsorbed on their surface, known as protein corona, which may alter the targeting ability of the nanoparticles. The impact of different protein corona on the "passive" and "active" targeting behaviors is still ambiguous. Herein, three kinds of aqueous soluble Fe3O4 nanoparticles with different surface modifications were synthesized and applied to explore the correlation between their protein corona and passive/active tumor-targeting abilities. In the in vitro and in vivo studies, the protein corona exhibited completely different effects on the active and passive cancer-targeting capability of the particles. The particles presented active cancer-targeting ability if there was enough interaction time between the particles and cells. This was mainly due to the dynamic evolution of the protein corona, the proteins of which may be outcompeted by the cancer cell membrane and determine the targeting abilities. Unfortunately, the protein corona also inevitably accelerated RES/MPS uptake after the particles were injected into the body, which almost completely disabled the active targeting abilities of the particles. We believe that this in-depth understanding of protein corona will provide new ideas on the tumor-targeting mechanisms of nanoparticles and present a feasible approach to designing targeted drugs in the future.


Assuntos
Antineoplásicos/farmacologia , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas
17.
Inorg Chem ; 61(3): 1456-1470, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-34995063

RESUMO

Indolo[2,3-d]benzazepines (indololatonduines) are rarely discussed in the literature. In this project, we prepared a series of novel indololatonduine derivatives and their RuII and OsII complexes and investigated their microtubule-targeting properties in comparison with paclitaxel and colchicine. Compounds were fully characterized by spectroscopic techniques (1H NMR and UV-vis), ESI mass-spectrometry, and X-ray crystallography, and their purity was confirmed by elemental analysis. The stabilities of the compounds in DMSO and water were confirmed by 1H and 13C NMR and UV-vis spectroscopy. Novel indololatonduines demonstrated anticancer activity in vitro in a low micromolar concentration range, while their coordination to metal centers resulted in a decrease of cytotoxicity. The preliminary in vivo activity of the RuII complex was investigated. Fluorescence staining and in vitro tubulin polymerization assays revealed the prepared compounds to have excellent microtubule-destabilizing activities, even more potent than the well-known microtubule-destabilizing agent colchicine.


Assuntos
Antineoplásicos/farmacologia , Complexos de Coordenação/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Indóis/farmacologia , Microtúbulos/efeitos dos fármacos , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Compostos Heterocíclicos com 3 Anéis/química , Humanos , Indóis/química , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Microtúbulos/metabolismo , Modelos Moleculares , Estrutura Molecular , Polimerização/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas
18.
Chem Commun (Camb) ; 58(14): 2315-2318, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35076033

RESUMO

Herein, we report the design of therapeutic nanoparticles by encapsulating photosensitizers and aluminum ions into metal-organic frameworks. The nanoparticles could significantly inhibit the growth of primary and rechallenged tumors by a combination of photothermal therapy and immunotherapy. This work offers a promising strategy to design an immunologic nanoplatform for "cold" tumor therapy.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia , Estruturas Metalorgânicas/farmacologia , Terapia Fototérmica , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Estruturas Metalorgânicas/síntese química , Estruturas Metalorgânicas/química , Camundongos , Estrutura Molecular , Tamanho da Partícula
19.
Chem Res Toxicol ; 35(2): 275-282, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35050609

RESUMO

Many chemotherapeutic drugs exert their cytotoxicity through the formation of DNA modifications (adducts), which interfere with DNA replication, an overactive process in rapidly dividing cancer cells. Side effects from the therapy are common, however, because these drugs also affect rapidly dividing noncancerous cells. Hypoxia-activated prodrugs (HAPs) have been developed to reduce these side effects as they preferentially activate in hypoxic environments, a hallmark of solid tumors. CP-506 is a newly developed DNA-alkylating HAP designed to exert strong activity under hypoxia. The resulting CP-506-DNA adducts can be used to elucidate the cellular and molecular effects of CP-506 and its selectivity toward hypoxic conditions. In this study, we characterize the profile of adducts resulting from the reaction of CP-506 and its metabolites CP-506H and CP-506M with DNA. A total of 39 putative DNA adducts were detected in vitro using our high-resolution/accurate-mass (HRAM) liquid chromatography tandem mass spectrometry (LC-MS3) adductomics approach. Validation of these results was achieved using a novel strategy involving 15N-labeled DNA. A targeted MS/MS approach was then developed for the detection of the 39 DNA adducts in five cancer cell lines treated with CP-506 under normoxic and hypoxic conditions to evaluate the selectivity toward hypoxia. Out of the 39 DNA adducts initially identified, 15 were detected, with more adducts observed from the two reactive metabolites and in cancer cells treated under hypoxia. The presence of these adducts was then monitored in xenograft mouse models bearing MDA-MB-231, BT-474, or DMS114 tumors treated with CP-506, and a relative quantitation strategy was used to compare the adduct levels across samples. Eight adducts were detected in all xenograft models, and MDA-MB-231 showed the highest adduct levels. These results suggest that CP-506-DNA adducts can be used to better understand the mechanism of action and monitor the efficacy of CP-506 in vivo, as well as highlight a new role of DNA adductomics in supporting the clinical development of DNA-alkylating drugs.


Assuntos
Adutos de DNA/análise , DNA Bacteriano/análise , DNA/análise , Hipóxia/tratamento farmacológico , Pró-Fármacos/química , Animais , Bovinos , Feminino , Humanos , Hipóxia/metabolismo , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Estrutura Molecular , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , Células Tumorais Cultivadas
20.
Int J Immunopathol Pharmacol ; 36: 20587384211059673, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35037503

RESUMO

Angiogenesis is the process of new vascular formation, which is derived from various factors. For suppressing cancer cell growth, targeting angiogenesis is one of the therapeutic approaches. Vascular endothelial growth factor family receptors, including Flt-1, Flk-1 and Flt-4, have been found to play an essential role in regulating angiogenesis. Rapamycin is a macrolide compound with anti-proliferative properties, while platelet factor-4 (PF-4) is an antiangiogenic ELR-negative chemokine. Rapamycin inhibits mTOR ligands activation, thus suppressing cell proliferation, while PF-4 inhibits cell proliferation through several mechanisms. In the present study, we evaluated the effects of rapamycin and platelet factor-4 toward breast carcinoma at the proteomic and genomic levels. A total of 60 N-Methyl-N-Nitrosourea-induced rat breast carcinomas were treated with rapamycin, platelet factor-4 and rapamycin+platelet factor-4. The tumours were subsequently subjected to immunohistochemical protein analysis and polymerase chain reaction gene analysis. Protein analysis was performed using a semiquantitative scoring method, while the mRNA expression levels were analysed based on the relative expression ratio. There was a significant difference in the protein and mRNA expression levels for the selected markers. In the rapamycin+platelet factor-4-treated group, the Flt-4 marker was downregulated, whereas there were no differences in the expression levels of other markers, such as Flt-1 and Flk-1. On the other hand, platelet factor-4 did not exhibit a superior angiogenic inhibiting ability in this study. Rapamycin is a potent antiangiogenic drug; however, platelet factor-4 proved to be a less effective drug of anti-angiogenesis on rat breast carcinoma model.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Fator Plaquetário 4/administração & dosagem , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Sirolimo/administração & dosagem , Animais , Feminino , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Metilnitrosoureia , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...