Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(3): e0299499, 2024.
Article in English | MEDLINE | ID: mdl-38527041

ABSTRACT

Chemo-immunotherapy uses combined systemic therapies for resectable and unresectable tumors. This approach is gaining clinical momentum, but survival increases leave considerable room for improvement. A novel form of Pulsed Electric Field (PEF) ablation combines focal tissue destruction with immune activation in preclinical settings. The PEFs induce lethal cell damage without requiring thermal processes, leaving cellular proteins intact. This affords PEF a favorable safety profile, improved antigenicity, and significant immunostimulatory damage-associated molecular pattern release compared to other focal therapies. Preclinical investigations demonstrate a combinatorial benefit of PEF with immunostimulation. This study evaluates whether this proprietary PEF therapy induces an immunostimulatory effect sufficient to augment systemic neoadjuvant chemotherapy and immunotherapy to reverse metastatic disease in an immune-cold murine tumor model. To determine whether PEF improves a neoadjuvant chemo-immunotherapy standard-of-care, partial PEF ablation was delivered to orthotopically inoculated 4T1 metastatic tumors in addition to combinations of cisplatin chemotherapy and/or αPD-1 immunotherapy, followed by resection. In addition, to determine whether PEF combined with chemo-immunotherapy improves local and metastatic response in unresectable populations, partial PEF ablation was added to chemo-immunotherapy in mice that did not receive resection. Blood cytokines and flow cytometry evaluated immune response. Partial PEF ablation generates an immunostimulatory tumor microenvironment, increases systemic immune cell populations, slows tumor growth, and prolongs survival relative to neoadjuvant systemic therapies-alone. These data suggest the addition of this proprietary PEF locoregional therapy may synergize with systemic standard-of-care paradigms to improve outcomes with potential or demonstrated metastatic disease in both resectable and unresectable patient cohorts.


Subject(s)
Neoadjuvant Therapy , Neoplasms , Mice , Humans , Animals , Disease Models, Animal , Neoplasms/pathology , Immunotherapy , Cisplatin/therapeutic use , Tumor Microenvironment
2.
J Vasc Interv Radiol ; 35(3): 442-451.e7, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38042523

ABSTRACT

PURPOSE: To compare the immune response and survival after size-matched radiofrequency (RF) ablation and a proprietary form of pulsed electric field (PEF) ablation in murine tumors. MATERIAL AND METHODS: Orthotopically inoculated EMT6 or 4T1 murine tumors received sham, RF ablation, or PEF ablation. 4T1 tumor ablations included subgroups with intraperitoneal checkpoint inhibition immunotherapy (αPD-1). Blood was collected for cytokine profiling and flow cytometry. Tumor size was measured and survival was monitored. Tumor samples were processed for histology, immunohistochemistry, flow cytometry, and cytokine profiling. Lungs were collected from 4T1-bearing mice for hematoxylin and eosin histology to assess metastatic spread and abscopal effect induced by ablation. RESULTS: PEF elicited distinct immunomodulatory effects, with clear differences in serum and tumor cytokine profiles compared with RF ablation, including intratumoral downregulation of vascular endothelial growth factor, hypoxia-inducible factor 1α, c-MET, interleukin-10, Ki67, and tumor necrosis factor-α (all P < .05). PEF increased innate immune activation, with enhanced recruitment of dendritic cells, M1 macrophages, and natural killer cells coupled with a reduction in M2 macrophages and myeloid-derived suppressor cells (all P < .05). Concurrently, PEF strengthened adaptive immunity compared with RF ablation, characterized by increased antigen-specific T cells and decreased regulatory T cells (all P < .05). PEF stalled tumor growth and increased survival at the end of the study (≥4× versus RFA). Finally, PEF promoted an abscopal effect of clearing metastases in the lungs, which was stronger in combination with αPD-1 than with PEF alone. CONCLUSIONS: The proprietary form of PEF used in this study evoked a preferential immunostimulatory profile versus RF ablation thermal ablation in mice, with implications for enhancing the therapeutic effectiveness of checkpoint inhibition immunotherapy for immunotherapy-unresponsive tumors.


Subject(s)
Neoplasms , Radiofrequency Ablation , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , T-Lymphocytes/metabolism , Cytokines/metabolism
3.
PLoS One ; 16(2): e0247652, 2021.
Article in English | MEDLINE | ID: mdl-33626096

ABSTRACT

Breast cancer is the leading cause of cancer-related deaths in the United States. The majority of deaths (90%) in breast cancer patients is caused by invasion and metastasis-two features related to the epithelial-to-mesenchymal transition (EMT). Twist1 is a key transcription factor that promotes the EMT, which leads to cell migration, invasion, cancer metastasis, and therapeutic resistance. Harmine is a beta-carboline alkaloid found in a variety of plants and was recently shown to be able to induce degradation of Twist Family BHLH Transcription Factor 1 (Twist1) in non-small cell lung cancer cells (NSCLC). In this study, we show that harmine can inhibit migration and invasion of both human and mouse breast cancer cells in a dose-dependent manner. Further study shows that this inhibition is most likely achieved by inducing a proteasome-dependent Twist1 degradation. At the concentrations tested, harmine did not affect the viability of cells significantly, suggesting that its inhibition of cancer cell migration and invasion is largely independent of its cytotoxicity, but due to its ability to affect regulators of EMT such as Twist1. This result may facilitate the development of strategies that target Twist1 to treat metastatic breast cancer, as Twist1 is expressed at a high level in metastatic breast cancer cells but not in normal cells.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Harmine/pharmacology , Proteolysis/drug effects , Twist-Related Protein 1/metabolism , Animals , Breast Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Humans , Mice , Neoplasm Invasiveness/pathology , Wound Healing/drug effects
4.
Drug Chem Toxicol ; 44(3): 286-293, 2021 May.
Article in English | MEDLINE | ID: mdl-30938206

ABSTRACT

Carbon tetrachloride (CCl4) is a strong hepatotoxic agent. The ability of the anti-inflammatory agent, lactoferrin (LF), to alleviate hepatic inflammation in a Wistar rat model administered with carbon tetrachloride (CCl4) was examined. Thirty male Wistar rats were segregated into 5 groups (6 rats per group): Control group, LF group (300 mg LF/kg b. wt daily for three weeks), CCl4 group (1 ml CCl4/kg b. wt once orally), LF-protected group (300 mg LF/kg b. wt daily for 3 weeks followed by 1 mL CCl4/kg b. wt once orally), and LF-treated group (1 mL CCl4/kg b.wt once orally followed by 300 mg LF/kg b. wt orally every day for three weeks). Erythrogram, leukogram, activity of oxidative stress markers (Superoxide dismutase [SOD], Glutathione peroxidase [GPx], and Malondialdehyde [MDA]), and expression of hepatic paraoxonase-1 (PON1), interleukin (IL)-1ß, and IL-10 mRNA were determined. Histopathological examination of the hepatic tissue was carried out. CCl4 caused liver injury, loss of liver antioxidant activity of SOD and GPx, and a significant increase in the level of malondialdehyde in the serum. Moreover, CCl4 induced up-regulation of hepatic pro-inflammatory (IL-1ß) factors, and down-regulation of anti-inflammatory (IL-10 and PON1) factors. Based on histopathological examination, the hepatic tissues had severe inflammation and were damaged. However, LF mitigated the liver damage, oxidative stress, and hepatotoxicity caused by CCl4. Overall, these results suggest that LF-mediated immunological mechanisms alleviate CCl4-induced hepatic toxicity and provide a novel perspective on the potential use of LF for prophylactic and therapeutic applications in treating liver diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Lactoferrin/pharmacology , Administration, Oral , Animals , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/pathology , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
5.
Curr Pharm Biotechnol ; 21(1): 23-36, 2020.
Article in English | MEDLINE | ID: mdl-31438827

ABSTRACT

BACKGROUND: Vitamin C (VC) is believed to enhance immunity and is regularly integrated as a supplementary agent during several treatments. OBJECTIVE: The green (GC) and roasted (RC) coffee (Coffea arabica) aqueous extracts (0, 125, 250 and 500 µg/ml) combined with VC (50 µg/ml) were examined on the cancerous MCF-7 cell line and normal human lymphocytes. METHODS: Neutral red uptake assay, comet assay, immunocytochemical reactivity for protein expression and mRNA expression of apoptosis-related genes were performed. RESULTS: A significant (P< 0.05) concentration-dependent increase of apoptotic features, such as morphological changes, and abundant nuclear condensation, altered the expression of p53 and caspase-3 mRNA, down-regulation of Bcl-2 protein as well as the acidic autophagosomal vacuolization in treated cells. The oxidative stress and DNA single-strand breaks were noticed too. CONCLUSION: These results suggest that coffee in combination with VC undergoes apoptotic anticancer pathway. This supports the integration of coffee and VC as a valuable candidate for anticancer research and treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Coffea , Plant Extracts/pharmacology , Vitamins/pharmacology , Cell Death/drug effects , Humans , MCF-7 Cells , Seeds
6.
Anticancer Agents Med Chem ; 19(9): 1103-1113, 2019.
Article in English | MEDLINE | ID: mdl-30914034

ABSTRACT

BACKGROUND: Fennel (Foeniculum vulgare) and rose geranium (Pelargonium graveolens) oils are known for their various biological effects including anticancer properties. OBJECTIVE: This study aimed to evaluate the anticancer mechanism of fennel and geranium oils combined treatment on MCF-7 cells. METHODS: The GC-MS method for essential oil characterization as well as the in vitro cytotoxicity, morphological changes, real-time PCR and immunocytochemical investigation for apoptosis-related markers, in addition, to flow cytometric cell cycle distribution analysis were done. RESULTS: The major constituents of both essential oils were anethole (55.33 %) and estragole (11.57 %) for fennel essential oil. However, cintronellol (34.40 %) and geraniol (8.67 %) were identified in geranium oil. The results revealed an IC50 of 220±5.7 and 60±2.1µg/ml for fennel and geranium oils, respectively. The mechanistic anticancer properties were investigated throughout the 70, 50, and 25µg/ml of oils mixture. The marked apoptotic morphology and the flow cytometric cell cycle distribution analysis in addition to the levels of apoptosisrelated makers such as p53, caspase-3, mir-21, mir-92a, Bcl-2, and ki-67 confirmed that fennel and geranium oils combination induced cell cycle arrest and apoptosis in MCF-7 cells. Moreover, the oils mixture did not exert any significant (P<0.01) toxicity on normal human peripheral blood lymphocytes in vitro. CONCLUSION: The findings showed that the mixture of oils exerted selective cytotoxicity towards MCF-7 cells through induction of cell cycle arrest and apoptosis which may be triggered by the synergistic effect between the active ingredients of fennel and geranium oils.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Foeniculum/chemistry , Oils, Volatile/pharmacology , Pelargonium/chemistry , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gas Chromatography-Mass Spectrometry , Humans , MCF-7 Cells , Molecular Structure , Oils, Volatile/chemistry , Oils, Volatile/isolation & purification , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Structure-Activity Relationship
7.
Int J Mol Sci ; 20(4)2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30781605

ABSTRACT

This study was designed to investigate the potential effects and underlying mechanism of adipose tissue-derived mesenchymal stem cells (MSCs) on allergic inflammation compared to Montelukast as an antileukotriene drug in a rat model of allergic rhinitis (AR). The effect of MSCs was evaluated in albino rats that were randomly divided into four (control, AR, AR + Montelukast, and AR + MSCs) groups. Rats of AR group were sensitized by ovalbumin (OVA) and then challenged with daily nasal drops of OVA diluted in sterile physiological saline (50 µL/nostril, 100 mg/mL, 10% OVA) from day 15 to day 21 of treatment with/without Montelukast (1 h before each challenge) or MSCs I/P injection (1 × 106 MCSs; weekly for three constitutive weeks). Both Montelukast and MSCs treatment started from day 15 of the experiment. At the end of the 5th week, blood samples were collected from all rats for immunological assays, histological, and molecular biology examinations. Both oral Montelukast and intraperitoneal injection of MSCs significantly reduced allergic symptoms and OVA-specific immunoglobulin E (IgE), IgG1, IgG2a and histamine as well as increasing prostaglandin E2 (PGE2). Further analysis revealed that induction of nasal innate cytokines, such as interleukin (IL)-4 and TNF-α; and chemokines, such as CCL11 and vascular cell adhesion molecule-1 (VCAM-1), were suppressed; and transforming growth factor-ß (TGF-ß) was up-regulated in Montelukast and MSCs-treated groups with superior effect to MSCs, which explained their underlying mechanism. In addition, the adipose tissue-derived MSCs-treated group had more restoring effects on nasal mucosa structure demonstrated by electron microscopical examination.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Rhinitis, Allergic/immunology , Rhinitis, Allergic/therapy , Animals , Cells, Cultured , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Disease Models, Animal , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Nasal Mucosa/pathology , Nasal Mucosa/ultrastructure , Rats , Rhinitis, Allergic/blood , Rhinitis, Allergic/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
8.
Anticancer Agents Med Chem ; 18(15): 2187-2192, 2018.
Article in English | MEDLINE | ID: mdl-30198441

ABSTRACT

BACKGROUND: Bone Marrow (BM) has the self-renovation capacity and has been used recently in tumor medicine. Chlorambucil [CHB] is ordinarily utilized chemotherapy to treat varieties of malignancy patients. This investigation intended to gauge the effectiveness of BM as an in-vivo antimutagenic against CHB. METHODS: The experimental design relies upon four classes; each class contains ten adult male albino rats as follows: control, rats infused orally with CHB for fourteen days, rats intravenously infused with BM through a tail vein one time, rats infused the mix of CHB and BM.The Anticancer capability of BM was assessed by cytogenetic assay and mitotic index. The declarations of the apoptosis- related genes were examined by RT-qPCR examination. RESULTS: The present experiment demonstrated a curative effect of BM against the cytotoxic impact of CHB. Infusion of BM after chemotherapy helps to diminish the chromosomal aberration; increment mitotic index and decline the Bax/Bcl2 proportion compared with [CHB] class gather that prompts expanding the survival rate of influenced cells with chemotherapy cytotoxicity. CONCLUSION: The present study shows that bone marrow transplantation together after CHB infusion helps to increase genomic stability by diminishing structural chromosome abnormalities, diminishing the Bax/Bcl2 proportion and increasing the mitotic index.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bone Marrow Cells/drug effects , Cell Survival/drug effects , Chlorambucil/pharmacology , Animals , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Chromosome Aberrations , Male , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics
9.
Nat Protoc ; 13(5): 819-839, 2018 05.
Article in English | MEDLINE | ID: mdl-29599440

ABSTRACT

This protocol is an extension to: Nat. Protoc. 6, 870-895 (2014); doi:10.1038/nprot.2011.328; published online 02 June 2011Cycling cells duplicate their DNA content during S phase, following a defined program called replication timing (RT). Early- and late-replicating regions differ in terms of mutation rates, transcriptional activity, chromatin marks and subnuclear position. Moreover, RT is regulated during development and is altered in diseases. Here, we describe E/L Repli-seq, an extension of our Repli-chip protocol. E/L Repli-seq is a rapid, robust and relatively inexpensive protocol for analyzing RT by next-generation sequencing (NGS), allowing genome-wide assessment of how cellular processes are linked to RT. Briefly, cells are pulse-labeled with BrdU, and early and late S-phase fractions are sorted by flow cytometry. Labeled nascent DNA is immunoprecipitated from both fractions and sequenced. Data processing leads to a single bedGraph file containing the ratio of nascent DNA from early versus late S-phase fractions. The results are comparable to those of Repli-chip, with the additional benefits of genome-wide sequence information and an increased dynamic range. We also provide computational pipelines for downstream analyses, for parsing phased genomes using single-nucleotide polymorphisms (SNPs) to analyze RT allelic asynchrony, and for direct comparison to Repli-chip data. This protocol can be performed in up to 3 d before sequencing, and requires basic cellular and molecular biology skills, as well as a basic understanding of Unix and R.


Subject(s)
Cell Division , DNA Replication , DNA/biosynthesis , High-Throughput Nucleotide Sequencing/methods , Mouse Embryonic Stem Cells/physiology , Time , Animals , Bromodeoxyuridine/metabolism , Cell Line , Chromatin Immunoprecipitation , Mice , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...