Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.352
Filter
1.
J Control Release ; 350: 803-814, 2022 10.
Article in English | MEDLINE | ID: mdl-36087802

ABSTRACT

Postsurgical treatment is of great importance to combat tumor recurrence and metastasis. Anti-CD47 antibodies (aCD47) can block the CD47-signal regulatory protein-alpha (CD47-SIRPα) pathway to restore immunity. Here, an in-situ gel implantation was engineered by crosslinking chitosan (CS) and pullulan (Pul) for postsurgical treatment. A highly selected chemotherapeutic, cyclopamine (Cyc), encapsulated in liposomes (Cyc-Lip) was co-loaded with aCD47 in gels for chemoimmunotherapy. Importantly, a sequential drug release kinetics can be achieved. Nanotherapeutics were confirmed to be released prior to aCD47 in a burst-release manner, which was benefit for immediately killing residual tumor cells followed by releasing tumor antigens. Meanwhile, aCD47 was released in a sustained-release manner to restore macrophage functions and exert anti-tumor immune responses. Afterwards, the efficacy of in-situ chemoimmunotherapy was confirmed on 4T1 mouse breast cancer models, which could not only efficiently augment anti-tumor effect to inhibit tumor recurrence but also establish a long-term immune memory to combat tumor metastasis.


Subject(s)
Anticarcinogenic Agents , Immunotherapy , Neoplasms , Postoperative Care , Animals , Anticarcinogenic Agents/administration & dosage , Antigens, Neoplasm , Chitosan/administration & dosage , Delayed-Action Preparations , Immunotherapy/methods , Mice , Neoplasm Recurrence, Local/prevention & control , Neoplasms/pathology , Neoplasms/surgery
2.
Bol. latinoam. Caribe plantas med. aromát ; 21(1): 66-80, ene. 2022. ilus, tab
Article in English | LILACS | ID: biblio-1372378

ABSTRACT

Melastoma malabathricum (M. malabathricum) extracts have been reported to exert various pharmacological activities including antioxidants, anti-inflammatory and antiproliferative activities. The objective of the present study was to determine the anticarcinogenic activity of its methanol extract (MEMM) against the azoxymethane (AOM)-induced early colon carcinogenesis in rats. Rats were randomly assigned to five groups (n=6) namely normal control, negative control, and treatment (50, 250 or 500 mg/kg of MEMM) groups. Colon tissues were harvested for histopathological analysis and endogenous antioxidant system determination. MEMM was also subjected to HPLC analysis. Findings showed that MEMM significantly (p<0.05) reversed the AOM-induced carcinogenicity by: i) reducing the formation of aberrant crypt foci (ACF) in colon tissues, and; ii) enhancing the endogenous antioxidant activity (catalase, superoxide dismutase and glutathione peroxidase). Moreover, various phenolics has been identified in MEMM. In conclusion, MEMM exerts the in vivo anticarcinogenic activity via the activation of endogenous antioxidant system and synergistic action of phenolics.


Se ha informado que los extractos de Melastoma malabathricum (M. malabathricum) ejercen diversas actividades farmacológicas, incluidas actividades antioxidantes, antiinflamatorias y antiproliferativas. El objetivo del presente estudio fue determinar la actividad anticancerígena de su extracto de metanol (MEMM) contra la carcinogénesis de colon temprana inducida por azoximetano (AOM) en ratas. Las ratas se asignaron al azar a cinco grupos (n=6), a saber, los grupos de control normal, control negativo y tratamiento (50, 250 o 500 mg/kg de MEMM). Tejidos de colon fueron recolectados para análisis histopatológico y determinación del sistema antioxidante endógeno. MEMM también se sometió a análisis de HPLC. Los hallazgos mostraron que MEMM invirtió significativamente (p<0.05) la carcinogenicidad inducida por AOM al: i) reducir la formación de focos de criptas aberrantes (ACF) en los tejidos del colon, y; ii) potenciar la actividad antioxidante endógena (catalasa, superóxido dismutasa y glutatión peroxidasa). Además, se han identificado varios fenólicos en MEMM. En conclusión, MEMM ejerce la actividad anticancerígena in vivo mediante la activación del sistema antioxidante endógeno y la acción sinérgica de los fenólicos.


Subject(s)
Animals , Rats , Plant Extracts/administration & dosage , Anticarcinogenic Agents/administration & dosage , Colonic Neoplasms/drug therapy , Melastomataceae/chemistry , Organ Size/drug effects , Body Weight/drug effects , Chromatography, High Pressure Liquid , Rats, Sprague-Dawley , Colon/pathology , Plant Leaves , Methanol , Phenolic Compounds , Aberrant Crypt Foci , Carcinogenesis/drug effects , Antioxidants
3.
Pharm Biol ; 59(1): 1490-1504, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34714214

ABSTRACT

CONTEXT: The development of nanocarriers of plant origin, such as plant cell membranes, has recently been investigated. Also, plant bioactive compounds as sulforaphane (SFN) from broccoli have recognized antioxidant or anticancer properties. OBJECTIVE: To investigate the capacity of membrane vesicles from broccoli (BM-vesicles) to encapsulate SFN and their application in the cancer cell line. MATERIALS AND METHODS: Physicochemical analysis was carried out to characterize BM-vesicles through different approaches: dynamic light scattering, transmission electron microscopy, stopped-flow analysis, and proteomic analysis. They were applied at different concentrations (BM-vesicles at 0.04-0.00315% of protein and SFN at 5, 25, and 100 µM) in SK-MEL-28 cells during 24 h for studying cytotoxicity and gene expression. RESULTS: The entrapment efficiency was 41%. The anticancer activity tested in cells showed a decrease in proliferation when SFN in BM-vesicles was utilized. Expression patterns when SFN was applied in an encapsulated form showed a reduction of cancer markers and an increase of AQP3. Also, the metabolism of SFN occurred inside of cells, and higher SFN penetrated when it was encapsulated. DISCUSSION: The results showed that encapsulated SFN was better absorbed by melanoma cells providing metabolism products and a reduction of cancer molecular markers. Also aquaporin, AQP3 was pointed to as an important marker since it appeared to play a key role in homeostasis due to the importance of water transport in biological processes. CONCLUSION: These results indicate that SFN and SFN encapsulated in BM-vesicles have a high activity for the inhibition of melanocyte development. Therefore, BM-vesicles could serve as nanocarriers for drugs.


Subject(s)
Anticarcinogenic Agents/pharmacology , Brassica/chemistry , Isothiocyanates/pharmacology , Melanoma/drug therapy , Sulfoxides/pharmacology , Anticarcinogenic Agents/administration & dosage , Aquaporin 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Isothiocyanates/administration & dosage , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/pathology , Nanocapsules , Proteomics , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Sulfoxides/administration & dosage
4.
Mol Pharm ; 18(9): 3401-3417, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34482696

ABSTRACT

The need of pharmacological strategies to preclude breast cancer development motivated us to develop a non-aqueous microemulsion (ME) capable of forming a depot after administration in the mammary tissue and uptake of interstitial fluids for prolonged release of the retinoid fenretinide. The selected ME was composed of phosphatidylcholine/tricaprylin/propylene glycol (45:5:50, w/w/w) and presented a droplet diameter of 175.3 ± 8.9 nm. Upon water uptake, the ME transformed successively into a lamellar phase, gel, and a lamellar phase-containing emulsion in vitro as the water content increased and released 30% of fenretinide in vitro after 9 days. Consistent with the slow release, the ME formed a depot in cell cultures and increased fenretinide IC50 values by 68.3- and 13.2-fold in MCF-7 and T-47D cells compared to a solution, respectively. At non-cytotoxic concentrations, the ME reduced T-47D cell migration by 75.9% and spheroid growth, resulting in ∼30% smaller structures. The depot formed in vivo prolonged a fluorochrome release for 30 days without producing any sings of local irritation. In a preclinical model of chemically induced carcinogenesis, ME administration every 3 weeks for 3 months significantly reduced (4.7-fold) the incidence of breast tumors and increased type II collagen expression, which might contribute to limit spreading. These promising results support the potential ME applicability as a preventive therapy of breast cancer.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Breast Neoplasms/prevention & control , Fenretinide/administration & dosage , Mammary Neoplasms, Experimental/prevention & control , Animals , Anticarcinogenic Agents/pharmacokinetics , Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation , Drug Screening Assays, Antitumor , Emulsions , Female , Fenretinide/pharmacokinetics , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea/administration & dosage , Methylnitrosourea/toxicity , Mice , Rats
5.
Drug Des Devel Ther ; 15: 4043-4052, 2021.
Article in English | MEDLINE | ID: mdl-34588765

ABSTRACT

INTRODUCTION: Synergy is defined as an interaction of some substances that cooperate to give rise to the combined effect greater than the sum of their individual effects. It is a natural strategy that has evolved by nature to more efficacy with low cost. METHODS: This study is designed to evaluate the chemopreventive effect of a combined drug sample which is prepared by mixing an equal portion of stigmasterol and palmatine isolated from Azadirachta indica and Tinospora cordifolia respectively at a concentration of 100 mg/kg and 200 mg/kg body weight during the whole concentration. RESULTS: At the end of the study, it was found that this combined drug sample decreased the number of tumors and their size. This drug significantly reduced the serum level of glutamate pyruvate transaminase, alkaline phosphatase, glutamate oxalate transaminase, and bilirubin and enhanced the level of oxidative enzyme level of glutathione, superoxide dismutase, and catalase, and inhibit the level of lipid peroxides. DISCUSSION: The result suggests that combined drug samples exhibit a chemopreventive effect which is better than the effect of individual drugs (stigmasterol and palmatine).


Subject(s)
Azadirachta/chemistry , Berberine Alkaloids/pharmacology , Stigmasterol/pharmacology , Tinospora/chemistry , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/isolation & purification , Anticarcinogenic Agents/pharmacology , Berberine Alkaloids/administration & dosage , Berberine Alkaloids/isolation & purification , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Female , Male , Mice , Stigmasterol/administration & dosage , Stigmasterol/isolation & purification
6.
Sci Rep ; 11(1): 13640, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34210998

ABSTRACT

Euglena gracilis is widely utilized as food or supplement to promote human and animal health, as it contains rich nutrients. In this study, we administered spray-dried powder of E. gracilis and paramylon, ß-glucan stored in E. gracilis cells, to A4gnt knockout (KO) mice. A4gnt KO mice are a mutant mouse model that spontaneously develops gastric cancer through hyperplasia-dysplasia-adenocarcinoma sequence in the antrum of the stomach, and we observed the effects of E. gracilis and paramylon on the early involvements of A4gnt KO mice. Male and female 10-week-old A4gnt KO mice and their age-matched wildtype C57BL/6J mice were orally administered with 50 mg of E. gracilis or paramylon suspended in saline or saline as a control. After 3-week administration, animals were euthanatized and the stomach was examined histopathologically and immunohistochemically. Gene expression patterns of the stomach, which have been reported to be altered with A4gnt KO, and IgA concentration in small intestine were also analyzed with real-time PCR and ELISA, respectively. Administration of Euglena significantly reduced the number of stimulated CD3-positive T-lymphocytes in pyloric mucosa of A4gnt KO mice and tend to reduce polymorphonuclear leukocytes infiltration. Euglena administration further downregulated the expression of Il11 and Cxcl1 of A4gnt KO mice. Euglena administration also affected IgA concentration in small intestinal contents of A4gnt KO mice. Paramylon administration reduced the number of CD3-positive lymphocytes in pyloric mucosa of A4gnt KO mice, and downregulated the expressions of Il11 and Ccl2 of A4gnt KO mice. Although we found no significant effects on gross and microscopic signs of gastric dysplasia and cell proliferation, the present study suggests that the administration of Euglena and paramylon may ameliorate the early involvements of A4gnt mice through the effects on inflammatory reactions in the gastric mucosa. The cancer-preventing effects should be studied with long-term experiments until actual gastric cancer formation.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Euglena gracilis , Glucans/therapeutic use , N-Acetylglucosaminyltransferases/genetics , Stomach Neoplasms/prevention & control , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/analysis , Dietary Supplements/analysis , Euglena gracilis/chemistry , Female , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Glucans/administration & dosage , Glucans/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
7.
Sci Rep ; 11(1): 6268, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737522

ABSTRACT

Perioperative systemic inflammation induced by surgical stress elevates the risk of hematogenous cancer metastasis. This study investigated the anti-metastatic effects and mechanisms of methylprednisolone (MP) administration for surgical stress. We examined the effects of MP on the expression of adhesion molecules in human vascular endothelial cells and in a murine hepatic metastasis model under lipopolysaccharide (LPS) administration, which mimics systemic inflammation induced by surgical stress. Serum E-selectin level was measured in blood samples obtained from 32 gastric cancer patients who were randomly assigned to treat preoperatively with or without MP. The expression of E-selectin in LPS-induced vascular endothelial cells was suppressed by MP. An adhesion assay showed the number of LPS-induced adherent tumour cells was significantly lower following MP. In the in vivo study, LPS significantly elevated the number of hepatic metastases, but pretreatment with MP before LPS significantly inhibited this elevation. The LPS-induced expression of E-selectin in the vascular endothelium of the portal vein was suppressed by MP. In human clinical samples, serum E-selectin level was significantly decreased by preoperative MP. Suppression of surgically induced systemic inflammation by MP administration might prevent hematogenous cancer metastases by suppressing the induction of E-selectin expression in the vascular endothelium.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Methylprednisolone/administration & dosage , Postoperative Complications/prevention & control , Stomach Neoplasms/blood , Stomach Neoplasms/surgery , Systemic Inflammatory Response Syndrome/prevention & control , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Disease Models, Animal , E-Selectin/blood , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/adverse effects , Liver Neoplasms/chemically induced , Male , Mice , Mice, Inbred BALB C , Postoperative Complications/blood , Preoperative Care/methods , Signal Transduction/drug effects , Stomach Neoplasms/pathology , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/chemically induced , Vascular Cell Adhesion Molecule-1/metabolism
8.
Toxicology ; 454: 152753, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33741493

ABSTRACT

The phytochemical sulforaphane (SF) has gained interest for its apparent association with reduced cancer risk and other cytoprotective properties, at least some of which are attributed to activation of the transcription factor Nrf2. Repair of bulky DNA adducts is important for mitigating carcinogenesis from exogenous DNA damaging agents, but it is unknown whether in vivo treatment with SF affects adduct repair. At 12 h following a single oral dose of 100 mg/kg SF, an almost doubling in activity for repair of pyridyloxobutylated DNA was observed in CD-1 mouse liver nuclear extracts, but not in lung extracts. This change at 12 h in repair activity was preceded by the induction of Nrf2-regulated genes but not accompanied by changes in levels of the specific nucleotide excision repair (NER) proteins XPC, XPA, XPB and p53 or in binding of hepatic XPC, XPA and XPB to damaged DNA. SF also did not significantly alter histone deacetylase activity as measured by acetylated histone H3 levels, or stimulate formation of γ-H2A.X, a marker of DNA damage. A significant reduction in oxidative DNA damage, as measured by 8-OHdG (a biomarker of oxidative DNA damage), was observed only in DNA from the lungs of SF-treated mice 3 h post-dosing. These results suggest that the ability of SF to increase bulky adduct repair activity is organ-selective and is consistent with activation of the Nrf2 signaling pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , Isothiocyanates/pharmacology , Sulfoxides/pharmacology , Animals , Anticarcinogenic Agents/administration & dosage , DNA Adducts/drug effects , Female , Isothiocyanates/administration & dosage , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Sulfoxides/administration & dosage
9.
Anticancer Res ; 41(3): 1299-1305, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33788721

ABSTRACT

BACKGROUND/AIM: A xanthophyll of fucoxanthin (Fx) is a potential chemopreventive agent. Familial adenomatous polyposis (FAP) is an inherited disease that is associated with a high risk of developing colorectal cancer. However, it remains unclear whether Fx can modify colorectal tumorigenesis in ApcMin/+ mice, a model mouse for human FAP. MATERIALS AND METHODS: We investigated the chemopreventive effect of Fx in dextran sodium sulfate (DSS)-treated ApcMin/+ mice. RESULTS: Administration of Fx in the diet for 5 weeks significantly suppressed the number of colorectal adenocarcinomas in DSS-treated male ApcMin/+ mice, although the treatment did not affect the occurrence of colorectal dysplastic crypts and adenoma in the mice. In addition, Fx down-regulated cyclin D1 expression (0.6-fold) in colorectal mucosa of ApcMin/+ mice when compared with that of the control mice. CONCLUSION: Fx possesses chemopreventive potential against progression of colorectal carcinogenesis in ApcMin/+ mice that receive inflammatory stimuli.


Subject(s)
Adenomatous Polyposis Coli/complications , Anticarcinogenic Agents/administration & dosage , Colorectal Neoplasms/prevention & control , Xanthophylls/administration & dosage , Animals , Colorectal Neoplasms/chemically induced , Cyclin D1/analysis , Cyclin D1/physiology , Dextran Sulfate , Disease Models, Animal , Male , Mice
10.
Chem Phys Lipids ; 235: 105057, 2021 03.
Article in English | MEDLINE | ID: mdl-33515592

ABSTRACT

A close link between cardiovascular diseases and cancer results from sharing the same modifiable risk factors (e.g. nutritional) and cardiotoxicity of anti-cancerous therapies. It justifies cardio-oncological preliminary studies on dietary factors, especially on those of possible anti-carcinogenic or cardioprotective properties. The main purpose was to evaluate the effect of pomegranate seed oil (PSO) and/or bitter melon extract (BME) supplementation of the diet of female rats suffering from mammary tumors on lipidomic profile (expressed as fatty acids, conjugated fatty acids (CFA), malondialdehyde (MDA), cholesterol and oxysterols content) of cardiac tissue. Total lipidomic profile and intensity of lipid peroxidation in hearts of DMBA-treated Sprague-Dawley rats and their healthy equivalents, both obtaining diet supplementation, were evaluated with different chromatographic techniques coupled with appropriate detection systems (GC-MS, GC-TOFMS, Ag+-HPLC-DAD, UF-HPLC-DAD). Dietary modifications neither diminished breast cancer incidence nor exerted explicit cardio-protective influence, however, they diminished cholesterol content, i.a. because of inhibition of the endogenous conversion of squalene to cholesterol in cardiac tissue. CFA were incorporated into cardiac tissue to a lesser extent in the cancerous process. PSO and BME anti-oxidant properties in pathological condition were only slightly reflected in MDA levels but not in oxysterols formation. Obtained results indicate considerable changes in dietary supplements' biological activity in pathological conditions and the need for clear distinction of drugs and dietary supplements, which is of utmost importance, especially for cancer survivors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/drug therapy , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/drug therapy , Oxysterols/metabolism , Plant Extracts/pharmacology , Plant Oils/pharmacology , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/chemistry , Breast Neoplasms/metabolism , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/chemistry , Cardiovascular Diseases/metabolism , Dietary Supplements , Disease Models, Animal , Female , Lipid Peroxidation/drug effects , Lipidomics , Momordica charantia/chemistry , Myocardium/metabolism , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Oils/administration & dosage , Plant Oils/chemistry , Pomegranate/chemistry , Rats , Rats, Sprague-Dawley
11.
Phytomedicine ; 81: 153432, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33310310

ABSTRACT

BACKGROUND: A natural pterostilbene analogue isolated from the herb Sphaerophysa salsula, 3'-hydroxypterostilbene (HPSB), exhibits antiproliferative activity in several cancer cell lines; however, the inhibitory effects of HPSB on skin carcinogenesis remains unclear. PURPOSE: The aim of this study was to evaluate the inhibitory effects of HPSB on two-stage skin carcinogenesis in mice and its potential mechanism. STUDY DESIGN AND METHODS: This study investigated the anti-inflammatory and anti-tumor effects of HPSB in the 12-O-tetradecanoylphorbol-13-acetate (TPA)-stimulated acute skin inflammation and 7,12-dimethylbenz[a]anthracene (DMBA)/TPA-induced two-stage skin carcinogenesis model. In addition, the effects of HPSB on the modulation of the phase I and phase II metabolizing enzymes in the DMBA-induced HaCaT cell model were investigated. RESULTS: The results provide evidence that topical treatment with HPSB significantly inhibits TPA-induced epidermal hyperplasia and leukocyte infiltration through the down-regulation of cyclooxygenase-2 (COX-2), matrix metalloprotein-9 (MMP-9), and ornithine decarboxylase (ODC) protein expression in mouse skin. Furthermore, HPSB suppresses DMBA/TPA-induced skin tumor incidence and multiplicity via the inhibition of proliferating cell nuclear antigen (PCNA), Cyclin B1 and cyclin-dependent kinase 1 (CDK1) expression in the two-stage skin carcinogenesis model. In addition, pretreatment with HPSB markedly reduces DMBA-induced cytochrome P450 1A1 (CYP1A1) and cytochrome P450 1B1 (CYP1B1) gene expression in human keratinocytes; however, HPSB does not significantly affect the gene expression of the phase II enzymes. CONCLUSION: This is the first study to show that topical treatment with HPSB prevents mouse skin tumorigenesis. Overall, our study suggests that natural HPSB may serve as a novel chemopreventive agent capable of preventing carcinogen activation and inflammation-associated tumorigenesis.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Anticarcinogenic Agents/pharmacology , Skin Neoplasms/prevention & control , Stilbenes/pharmacology , Tetradecanoylphorbol Acetate/toxicity , Administration, Topical , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/administration & dosage , Carcinogens/toxicity , Cyclooxygenase 2/metabolism , Drug Eruptions/etiology , Drug Eruptions/prevention & control , Female , Gene Expression Regulation/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Mice, Inbred ICR , Ornithine Decarboxylase/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Stilbenes/administration & dosage
12.
Environ Toxicol ; 36(4): 460-471, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33156559

ABSTRACT

BACKGROUND: Neferine (NEF) is nontoxic, bisbenzylisoquinoline alkaloid is derived from the seed embryo of lotus, a familiar medicinal plant. Although several mechanisms have been planned, an evident antitumor action pathway of NEF on the oral tumor is still not known. In the current study, we aimed at investigating the protecting effect of NEF against experimental oral carcinoma and clarify its possible mechanism through the induction of apoptosis, proliferation, and inflammatory signaling pathways. METHODS: The experimental hamsters were divided into four groups (I-IV) containing six hamsters each. The group I was control group, group II and III hamsters treated with 7,12-dimethylbenz(a)anthracene (DMBA) (0.5%) alone, thrice in a week for 10 weeks, and group III and IV hamsters received oral supplementation of NEF at a concentration of 15 mg/kg bw. All the hamsters were sacrificed after 16 weeks. RESULTS: Our results revealed that DMBA treated hamsters exhibited 100% oral tumor cell formation with high-tumor incidence (TI), tumor number (TN), tumor volume (TV), decreased levels of antioxidants, increased status of lipid peroxidation (LPO), and modulated the activities of liver marker agents as well as NF-kB, cell proliferation (PCNA), and p53 proteins. NEF supplementation in DMBA treated hamsters, resulted in delayed lesion synthesis, and brought back the levels of the biochemical parameters. In addition, immunostaining of NF-kB, PCNA, and p53 showed that they were inhibited by NEF. CONCLUSION: Thus, NEF might be considered a better chemopreventive drug in an experimental model of home-based primary care (HBPC). More research is necessary to study other pathways implicated in oral carcinomas and their modulation by NEF.


Subject(s)
Anticarcinogenic Agents/pharmacology , Benzylisoquinolines/pharmacology , Carcinogenesis/drug effects , Carcinoma, Squamous Cell/prevention & control , Drugs, Chinese Herbal/pharmacology , Mouth Neoplasms/prevention & control , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Anticarcinogenic Agents/administration & dosage , Antioxidants/metabolism , Apoptosis/drug effects , Benzylisoquinolines/administration & dosage , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cricetinae , Drugs, Chinese Herbal/administration & dosage , Epithelial Cells/drug effects , Lipid Peroxidation/drug effects , Male , Mouth Neoplasms/pathology , NF-kappa B/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
13.
Life Sci ; 265: 118827, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33253720

ABSTRACT

BACKGROUND: Most hepatocellular carcinoma cases are diagnosed at late stages of the disease, which makes it the second cause of cancer mortality worldwide. For advanced-stage patients, chemotherapeutic drugs are the best treatment option; however, their adverse effects and high cost are still major obstacles for effective treatment. Spirulina microalga is a rich source of nutritional and bioactive elements and potential pharmaceuticals, which has an -proliferative effect against several cancer cell lines. It also has a prophylactic effect against the early stages of some cancer models, including hepatocellular carcinoma. AIMS: The present study was carried out to evaluate the therapeutic anticarcinogenic effect of spirulina against advanced murine hepatocellular carcinoma. MAIN METHODS: Hepatocarcinoma was induced by a single injection of diethylnitrosamine (100 mg/kg, intraperitoneally) followed by 22 weekly injections of carbon-tetrachloride (0.5 mg/kg, i.p). Spirulina (250 and 500 mg/kg bw) was given orally, from week 25 to 28, after the establishment of hepatocellular carcinoma. KEY FINDINGS: Spirulina inhibited HCC structural and functional alterations, manifested by improving the survival rate, significantly decreasing the tumor marker AFP, and the count and size of the hepatic nodules, as well as downstaging HCC. This was accompanied with the augmentation of the endogenous antioxidant capacity, apoptosis (Bax) and the tumor suppressor protein (p53), as well as the suppression of tissue levels of the lipid peroxidation marker (MDA) and neoangiogenesis marker (VEGF). SIGNIFICANCE: In conclusion, spirulina has an anticarcinogenic effect against advanced hepatocellular carcinoma exerted through activating the tumor suppressor protein p53 and apoptosis, and suppressing oxidative stress and angiogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Hepatocellular/prevention & control , Liver Neoplasms/prevention & control , Spirulina/chemistry , Animals , Anticarcinogenic Agents/administration & dosage , Antioxidants/metabolism , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Diethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Liver Neoplasms/pathology , Male , Mice , Neovascularization, Pathologic/prevention & control , Oxidative Stress/drug effects , Survival Rate , Tumor Suppressor Protein p53/metabolism
14.
Int J Nanomedicine ; 15: 8217-8230, 2020.
Article in English | MEDLINE | ID: mdl-33122907

ABSTRACT

PURPOSE: Black raspberries (BRBs) and their anthocyanin-rich hydrophilic fractions (BRB-H) have exhibited significant chemopreventative activity across aerodigestive cancers. Lutein, the primary component of the BRB lipophilic fraction (BRB-L), also demonstrates bioactivity potential, but is less well characterized, in part because of its poor, innate bioavailability. For these lipophilic compounds to be accurately evaluated for anticancer efficacy, it is necessary to increase their functional bioavailability using delivery vehicles. Lutein has been delivered in commercial settings in emulsion form. However, emulsions are unstable, particularly in the gastrointestinal tract, which limit their use as an oral nutraceutical. Here, we evaluated lutein encapsulation and cellular uptake for nanoparticle (NP) delivery vehicles composed of three different materials synthesized via two different approaches. METHODS: Specifically, NPs were synthesized via smaller scale batch interfacial instability (II) sonication and semi-continuous high throughput electrohydrodynamic-mediated mixing nanoprecipitation (EM-NP) methods using polystyrene-polyethylene oxide (PSPEO) or polycaprolactone-polyethylene glycol (PCLPEG) block copolymers and PHOSPHOLIPON 90G® (P90G, Lipoid GmbH) lipids. Size distribution, lutein encapsulation efficiency (EE), and cellular uptake and delivery were evaluated for each NP formulation. RESULTS: NPs produced via high throughput EM-NP had higher EEs than NPs produced via batch II sonication, and P90G had the greatest EE (55%) and elicited faster cellular uptake in premalignant oral epithelial cells (SCC83) compared to other delivery systems. CONCLUSION: These qualities suggest P90G could be a beneficial candidate for future lutein in vitro delivery research and clinical translation for oral cancer prevention.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Lutein/administration & dosage , Nanoparticles/chemistry , Nanotechnology/methods , Polymers/chemistry , Anticarcinogenic Agents/pharmacology , Cell Line , Drug Delivery Systems , Humans , Hydrophobic and Hydrophilic Interactions , Lutein/pharmacology , Micelles , Nanoparticles/administration & dosage , Particle Size , Polyesters , Polyethylene Glycols , Precancerous Conditions/drug therapy , Precancerous Conditions/pathology
15.
Food Funct ; 11(11): 9634-9650, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33048099

ABSTRACT

Despite that colorectal cancer (CRC) is a severe global health problem, effective chemopreventive strategies against CRC are still lacking. Huang-qin tea (HQT), a healthy herbal tea, is prepared from the aerial parts of Scutellaria baicalensis Georgi and has been consumed in China for thousands of years. HQT contains abundant flavonoids, which display potent anticancer effects, but no research studies have investigated the cancer-preventive effects of HQT on CRC in vivo. Here, we found that HQT inhibits azoxymethane-induced aberrant crypt foci (ACF) formation in a preneoplastic colonic ACF rat model. The essential role of the gut microbiota in the chemopreventive effect of HQT on CRC in a pseudo-germ-free rat model was confirmed. Besides, HQT modulates inflammatory cytokine expression by significantly decreasing IL-1ß, IL-6, IL-10, and TNF-α expression, and elevating IFN-γ production. 16S rDNA sequencing analysis indicated that HQT regulated the gut microbiota by increasing the abundance of beneficial bacteria (Lachnoclostridium, Alistipes, Roseburia, and Lactococcus) and reducing the levels of Bacteroides, Parasutterella, and unidentified_Clostridiales. Fecal metabolomics showed that HQT modulated the AOM-induced metabolomic disorder, and these altered metabolites were almost involved in the lipid metabolic pathways. The Spearman correlation analysis revealed a correlation between the gut microbiota and fecal metabolites. Collectively, these results suggested that HQT exerted beneficial effects on host health by inhibiting inflammation, and by regulating the gut microbiota profile and certain metabolic pathways. In conclusion, HQT inhibits AOM-induced ACF formation by modulating the gut microbiota composition and improving metabolomic disorders, indicating the potential of HQT as a functional beverage candidate for the prevention and treatment of CRC.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Colonic Neoplasms/prevention & control , Functional Food , Scutellaria baicalensis , Tea , Aberrant Crypt Foci/chemically induced , Aberrant Crypt Foci/prevention & control , Animals , Azoxymethane , Colonic Neoplasms/chemically induced , Disease Models, Animal , Male , Rats , Rats, Wistar
16.
Life Sci ; 261: 118348, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32860803

ABSTRACT

AIMS: 3,3'-Diindolylmethane (DIM) has limited anti-cancer effects in gastric cancer. Hydrogen sulfide (H2S) plays an important role in the tumor development and therapy, cystathionine-ß-synthase (CBS) and cystathionine-γ-lyase (CSE), two key endogenous H2S biosynthesis enzymes, can affect endogenous H2S levels and alter cancer treatment. Our main objective was to investigate whether the aminooxyacetic acid (AOAA) and DL-Propargylglycine (PAG), two specific inhibitors of CBS and CSE, could assist DIM to exert a stronger anti-cancer effects in gastric cancer BGC-823 and SGC-7901 cells. MATERIALS AND METHODS: Cell proliferation was assayed by MTT and cell colony-forming assay. Apoptosis and migration were detected by Hoechst staining and scratch test respectively. Western blot was used to evaluate the expression of proteins related to proliferation, apoptosis and migration. KEY FINDINGS: Combination of AOAA or PAG with DIM synergistically inhibited proliferation and migration, increased apoptosis in gastric cancer cells. The p38-p53 axis was also further activated by the combination of AOAA or PAG with DIM. Exogenous H2S from sodium hydrosulfide, attenuated the efficacy of DIM in cancer cells by reducing the activation level of p38-p53 axis. Taken together, AOAA or PAG inhibited the expression of endogenous H2S biosynthesis enzymes and effectively enhanced susceptibility of gastric cancer to DIM through activating p38-p53 axis. SIGNIFICANCE: The current study highlight more precise requirements for the clinical application of sulfur-containing anti-cancer drugs, and open a new way to enhance the sensitivity of DIM in chemotherapy of gastric cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Hydrogen Sulfide/antagonists & inhibitors , Indoles/pharmacology , Stomach Neoplasms/drug therapy , Alkynes/administration & dosage , Alkynes/pharmacology , Aminooxyacetic Acid/administration & dosage , Aminooxyacetic Acid/pharmacology , Anticarcinogenic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Drug Synergism , Glycine/administration & dosage , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Hydrogen Sulfide/metabolism , Indoles/administration & dosage , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Int J Mol Sci ; 21(16)2020 Aug 16.
Article in English | MEDLINE | ID: mdl-32824281

ABSTRACT

The development of nanocomposites has swiftly changed the horizon of drug delivery systems in defining a new platform. Major understanding of the interaction of nanocomposites with cells and how the interaction influences intracellular uptake is an important aspect to study in order to ensure successful utilisation of the nanocomposites. Studies have suggested that the nanocomposites' ability to permeate into biological cells is attributable to their well-defined physicochemical properties with nanoscale size, which is relevant to the nanoscale components of biology and cellular organelles. The functionalized graphene oxide coated with polyethylene glycol, loaded with protocatechuic acid and folic acid (GOP-PCA-FA) nanocomposite intracellular uptake was analysed using transmission electron microscope. The accumulation of fluorescent-labelled nanocomposites in the HepG2 cell was also analysed using a fluorescent microscope. In vitro cellular uptake showed that there was uptake of the drug from 24 h into the cells and the release study using fluorescently tagged nanocomposite demonstrated that release and accumulation were observed at 24 h and 48 h. Moreover, the migration ability of tumor cells is a key step in tumor progression which was observed 48 h after treatment. The GOP serves as a potential nanocarrier system which is capable of improving the therapeutic efficacy of drugs and biomolecules in medical as well as pharmaceutical applications through the enhanced intracellular release and accumulation of the encapsulated drugs. Nonetheless, it is essential to analyse the translocation of our newly developed GOP-PCA-FA, and its efficiency for drug delivery, effective cellular uptake, and abundant intracellular accumulation would be compromised by possible untoward side effects.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Folic Acid/administration & dosage , Graphite/chemistry , Hydroxybenzoates/administration & dosage , Liver Neoplasms/metabolism , Nanoparticles/metabolism , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/chemistry , Biological Transport , Drug Liberation , Folic Acid/chemistry , Hep G2 Cells , Humans , Hydroxybenzoates/chemistry , Nanocomposites/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Vitamins/administration & dosage , Vitamins/chemistry
18.
Nutr Res ; 81: 19-37, 2020 09.
Article in English | MEDLINE | ID: mdl-32828967

ABSTRACT

Chlorophyll, a phytochemical responsible for the green pigmentation in plants, has been studied for almost 100 years for its biological activities in humans. Over the past 30 years, the potential chemopreventative activities of both natural chlorophylls and their processed induced derivatives as well as the semisynthetic forms, such as sodium copper chlorophyllin, have been the focus of many research efforts. Established as potential chemopreventative agents with little to no bioavailability themselves, the activities of chlorophyll derivatives were generally ascribed to their ability to modulate mutagen/carcinogen bioavailability, their metabolism, and ultimately their ability to decrease the "exposure" to these carcinogens for humans at risk. More recently, systemic activities of chlorophyll derivatives have been reported to include modulation of oxidative stress and regulation of xenobiotic metabolizing systems and gene expression of systems critical to prevention of initiation and/or progression of cancer including NFE2-related factor 2, nuclear factor kappa B, TGF-ß, and ß-catenin pathways. With this in mind, the goals of this review are to provide an update to the comprehensive review of Ferruzzi and Blakeslee (2007) to include new insights into the behavior of chlorophyll derivatives in the gut as well as evidence of the systemic bioavailability of chlorophyll derivatives and their metabolites in support of potential impacts in prevention of cancer throughout the body.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Chlorophyll/analogs & derivatives , Diet , Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/pharmacology , Biological Availability , Carcinogens/metabolism , Carcinogens/pharmacokinetics , Chemoprevention , Chlorophyll/administration & dosage , Chlorophyll/metabolism , Chlorophyll/pharmacokinetics , Digestion , Digestive System/metabolism , Humans , Intestinal Absorption , Mutagens/metabolism , Oxidative Stress , Signal Transduction , Xenobiotics/metabolism
19.
J Trace Elem Med Biol ; 62: 126549, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32731109

ABSTRACT

BACKGROUND: Scientists are working on creating novel materials that can help in the treatment of diverse cancer-related diseases having trademark highlights like the target siting, specificity, improved therapeutic index of radiotherapy and chemotherapeutic treatments. The utilization of novel nanomaterials which are surface adorned with drugs or natural compounds can be used in diverse medical applications and helps in setting up a new platform for its improvement in the chemotherapeutic potentiality. One such nanomaterial is the trace element selenium in its nanoparticulate form that has been proved to be a potential chemotherapeutic agent recently. METHODS: The English language papers were gathered from electronic databases like Sciencedirect, Pub Med, Google Scholar and Scopus, the papers are published from 2001 to 2019. RESULTS: In the initial phase, approximately 200 papers were searched upon, out of which 118 articles were included after screening and critical reviewing. The information included was also tabulated for better knowledge and easy read. These articles contain information on the nanotechnology, inflammation, cancer and selenium as nanoparticles. CONCLUSION: The overview of the paper explains the enhancement of potentiality of anticancer drugs or phytochemicals which restricts its utilization in chemotherapeutic applications by the encapsulation or adsorption of them on selenium nanoparticles proven to accelerate the anticancerous properties with better results when compared with individual components. SeNPs (selenium nanoparticles) have demonstrated chemotherapeutic activity due to pro-oxidant property, where the anti-oxidant enzymes are stimulated to produce reactive active species, which induces oxidative stress, followed by activation of the apoptotic signalling pathway, cell cycle arrest, mitochondrial dysfunction and other pathways that ultimately lead to cell death. Selenium in nanoparticulate form can be used as a micronutrient to human health, thereby having low toxicity, can easily be degraded and also has good biocompatibility.


Subject(s)
Anticarcinogenic Agents/pharmacology , Nanoparticles/administration & dosage , Selenium/pharmacology , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Drug Delivery Systems/methods , Humans , Inflammation/drug therapy , Nanoparticles/chemistry , Nanoparticles/toxicity , Nanotechnology/methods , Oxidative Stress/drug effects , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/pharmacology , Selenium/administration & dosage , Selenium/chemistry
20.
Int J Pharm ; 586: 119475, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32525080

ABSTRACT

The purpose of this study was to develop solid dispersions of fenretinide(4HPR), incorporate them into poly(lactic-co-glycolic)(PLGA) millicylindrical implants, and evaluate the resulting implants in vitro and in vivo for future applications in oral cancer chemoprevention. Due to the extreme hydrophobicity of 4HPR, 4HPR-polyvinylpyrrolidone (PVP) amorphous solid dispersions(ASDs) were prepared for solubility enhancement. The optimal PVP-4HPR ratio of 9/1(w/w) provided a 50-fold solubility enhancement in aqueous media, which was sustained over 1 week. PVP-4HPR ASD particles were loaded into PLGA millicylinders and drug release was evaluated in vitro in PBST and in vivo by recovery from subcutaneous injection in rats. While initial formulations of PLGA PVP-4HPR millicylinders only released 10% 4HPR in vitro after 28 days, addition of the plasticizer triethyl-o-acetyl-citrate(TEAC) into PVP-4HPR ASDs resulted in a 5.6-fold total increase in drug release. Remarkably, the TEAC-PVP-4HPR PLGA implants demonstrated slow, continuous, and nearly complete release over 1 month in vivo compared to a 25% release for our previously reported formulation incorporating solubilizers and pore-forming agents. Hence, a combination of PLGA plasticizer and ASD formation provides an avenue for long-term controlled release in vivo for the exceptionally difficult drug to formulate, 4HPR, and a suitable formulation for future evaluation in rodent models of oral cancer.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Drug Carriers/chemistry , Fenretinide/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Anticarcinogenic Agents/chemistry , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Compounding , Drug Liberation , Fenretinide/chemistry , Hydrophobic and Hydrophilic Interactions , Male , Mouth Neoplasms/drug therapy , Povidone/chemistry , Rats , Rats, Sprague-Dawley , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...