Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancer Med ; 13(9): e7212, 2024 May.
Article in English | MEDLINE | ID: mdl-38686626

ABSTRACT

BACKGROUND: A phase I clinical study for patients with locally advanced H&N cancer with a new class of botanical drug APG-157 provided hints of potential synergy with immunotherapy. We sought to evaluate the efficacy of the combination of APG-157 and immune checkpoint inhibitors. METHODS: CCL23, UM-SCC1 (human), and SCCVII (HPV-), MEER (HPV+) (murine) H&N cancer cell lines were utilized for in vitro and in vivo studies. We measured tumor growth by treating the mice with APG-157, anti-PD-1, and anti-CTLA-4 antibody combinations (8 groups). The tumor microenvironments were assessed by multi-color flow cytometry, immunohistochemistry, and RNA-seq analysis. Fecal microbiome was analyzed by 16S rRNA sequence. RESULTS: Among the eight treatment groups, APG-157 + anti-CTLA-4 demonstrated the best tumor growth suppression (p = 0.0065 compared to the control), followed by anti-PD-1 + anti-CTLA-4 treatment group (p = 0.48 compared to the control). Immunophenotype showed over 30% of CD8+ T cells in APG-157 + anti-CTLA-4 group compared to 4%-5% of CD8+ T cells for the control group. Differential gene expression analysis revealed that APG-157 + anti-CTLA-4 group showed an enriched set of genes for inflammatory response and apoptotic signaling pathways. The fecal microbiome analysis showed a substantial difference of lactobacillus genus among groups, highest for APG-157 + anti-CTLA-4 treatment group. We were unable to perform correlative studies for MEER model as there was tumor growth suppression with all treatment conditions, except for the untreated control group. CONCLUSIONS: The results indicate that APG-157 and immune checkpoint inhibitor combination treatment could potentially lead to improved tumor control.


Subject(s)
CTLA-4 Antigen , Head and Neck Neoplasms , Immune Checkpoint Inhibitors , Tumor Microenvironment , Animals , Mice , CTLA-4 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Cell Line, Tumor , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Female , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Disease Models, Animal
2.
Cancer Res Commun ; 4(3): 706-722, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38421310

ABSTRACT

Gigaxonin is an E3 ubiquitin ligase that plays a role in cytoskeletal stability. Its role in cancer is not yet clearly understood. Our previous studies of head and neck cancer had identified gigaxonin interacting with p16 for NFκB ubiquitination. To explore its role in cancer cell growth suppression, we analyzed normal and tumor DNA from cervical and head and neck cancers. There was a higher frequency of exon 8 SNP (c.1293 C>T, rs2608555) in the tumor (46% vs. 25% normal, P = 0.011) pointing to a relationship to cancer. Comparison of primary tumor with recurrence and metastasis did not reveal a statistical significance. Two cervical cancer cell lines, ME180 and HT3 harboring exon 8 SNP and showing T allele expression correlated with higher gigaxonin expression, reduced in vitro cell growth and enhanced cisplatin sensitivity in comparison with C allele expressing cancer cell lines. Loss of gigaxonin expression in ME180 cells through CRISPR-Cas9 or siRNA led to aggressive cancer cell growth including increased migration and Matrigel invasion. The in vitro cell growth phenotypes were reversed with re-expression of gigaxonin. Suppression of cell growth correlated with reduced Snail and increased e-cadherin expression. Mouse tail vein injection studies showed increased lung metastasis of cells with low gigaxonin expression and reduced metastasis with reexpression of gigaxonin. We have found an association between C allele expression and RNA instability and absence of multimeric protein formation. From our results, we conclude that gigaxonin expression is associated with suppression of epithelial-mesenchymal transition through inhibition of Snail. SIGNIFICANCE: Our results suggest that GAN gene exon 8 SNP T allele expression correlates with higher gigaxonin expression and suppression of aggressive cancer cell growth. There is downregulation of Snail and upregulation of e-cadherin through NFκB ubiquitination. We hypothesize that exon 8 T allele and gigaxonin expression could serve as diagnostic markers of suppression of aggressive growth of head and neck cancer.


Subject(s)
Head and Neck Neoplasms , Humans , Animals , Mice , Down-Regulation/genetics , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/genetics , Cadherins/genetics
3.
OTO Open ; 6(3): 2473974X221109838, 2022.
Article in English | MEDLINE | ID: mdl-35860617

ABSTRACT

Inhalant toxicants are postulated to contribute to the pathogenesis of chronic rhinosinusitis. Permethrin is a pesticide widely used in agricultural, industrial, and residential settings. The objective of this pilot study is to investigate the in vitro effects of permethrin on sinonasal epithelial cells (SNECs). Sinus mucosa was collected from 4 patients undergoing transsphenoidal pituitary surgery without a history of chronic rhinosinusitis. Cultured SNECs were exposed to varied concentrations of permethrin (0-156 µM) for 6 days. Cell viability and proliferation were determined via the MTT colorimetric assay and the Incucyte Live Cell Imaging System. Cellular reactive oxygen species (ROS) activity was measured by the DCFDA ROS detection assay. A statistically significant reduction in cell viability and proliferation was observed between the exposure and control groups at certain concentrations, and a dose-dependent increase in ROS activity was also observed. These findings indicate that permethrin may have deleterious effects on SNECs in a dose-dependent manner.

4.
Front Oncol ; 12: 869108, 2022.
Article in English | MEDLINE | ID: mdl-35600369

ABSTRACT

Liquid biopsies are gaining more traction as non-invasive tools for the diagnosis and monitoring of cancer. In a new paradigm of cancer treatment, a synergistic botanical drug combination (APG-157) consisting of multiple molecules, is emerging as a new class of cancer therapeutics, targeting multiple pathways and providing a durable clinical response, wide therapeutic window and high level of safety. Monitoring the efficacy of such drugs involves assessing multiple molecules and cellular events simultaneously. We report, for the first time, a methodology that uses circulating plasma cell-free RNA (cfRNA) as a sensitive indicator of patient response upon drug treatment. Plasma was collected from six patients with head and neck cancer (HNC) and four healthy controls receiving three doses of 100 or 200 mg APG-157 or placebo through an oral mucosal route, before treatment and on multiple points post-dosing. Circulating cfRNA was extracted from plasma at 0-, 3- and 24-hours post-treatment, followed by RNA sequencing. We performed comparative analyses of the circulating transcriptome and were able to detect significant perturbation following APG-157 treatment. Transcripts associated with inflammatory response, leukocyte activation and cytokine were upregulated upon treatment with APG-157 in cancer patients, but not in healthy or placebo-treated patients. A platelet-related transcriptional signature could be detected in cancer patients but not in healthy individuals, indicating a platelet-centric pathway involved in the development of HNC. These results from a Phase 1 study are a proof of principle of the utility of cfRNAs as non-invasive circulating biomarkers for monitoring the efficacy of APG-157 in HNC.

5.
OTO Open ; 5(2): 2473974X211009232, 2021.
Article in English | MEDLINE | ID: mdl-34017935

ABSTRACT

Although the etiology of chronic rhinosinusitis remains unknown, environmental factors including airborne pollutants and toxicants are postulated to contribute to its pathogenesis. However, the precise pathomechanisms with which environmental toxicants may contribute to chronic rhinosinusitis are not fully understood. The purpose of this pilot study is to examine the cytotoxic effects of N,N-diethyl-meta-toluamide (DEET), a commonly used pesticide, on sinonasal epithelial cells (SNECs). Sinus mucosa was obtained from 3 subjects without a history of chronic rhinosinusitis. Cultured SNECs were exposed to various concentrations of DEET (0-5 mM) for 6 days. Cell viability, proliferation, and morphologic changes were assessed using the MTT colorimetric dye assay and the Incucyte Live Cell Monitoring System. Statistically significant dose-dependent reduction in cell viability and proliferation was observed between exposure and control groups (P < .05) at all concentrations tested. Dose-dependent cellular morphological changes were also seen. These findings indicate that DEET exposure induces dose-dependent cytotoxicity in sinonasal epithelia.

6.
J Biol Chem ; 295(50): 17169-17186, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33028635

ABSTRACT

We have observed overexpression of PACS-1, a cytosolic sorting protein in primary cervical tumors. Absence of exonic mutations and overexpression at the RNA level suggested a transcriptional and/or posttranscriptional regulation. University of California Santa Cruz genome browser analysis of PACS-1 micro RNAs (miR), revealed two 8-base target sequences at the 3' terminus for hsa-miR-34a and hsa-miR-449a. Quantitative RT-PCR and Northern blotting studies showed reduced or loss of expression of the two microRNAs in cervical cancer cell lines and primary tumors, indicating dysregulation of these two microRNAs in cervical cancer. Loss of PACS-1 with siRNA or exogenous expression of hsa-miR-34a or hsa-miR-449a in HeLa and SiHa cervical cancer cell lines resulted in DNA damage response, S-phase cell cycle arrest, and reduction in cell growth. Furthermore, the siRNA studies showed that loss of PACS-1 expression was accompanied by increased nuclear γH2AX expression, Lys382-p53 acetylation, and genomic instability. PACS-1 re-expression through LNA-hsa-anti-miR-34a or -449a or through PACS-1 cDNA transfection led to the reversal of DNA damage response and restoration of cell growth. Release of cells post 24-h serum starvation showed PACS-1 nuclear localization at G1-S phase of the cell cycle. Our results therefore indicate that the loss of hsa-miR-34a and hsa-miR-449a expression in cervical cancer leads to overexpression of PACS-1 and suppression of DNA damage response, resulting in the development of chemo-resistant tumors.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm , MicroRNAs/metabolism , RNA, Neoplasm/metabolism , Uterine Cervical Neoplasms/metabolism , Vesicular Transport Proteins/metabolism , Female , G1 Phase , HeLa Cells , Humans , MicroRNAs/genetics , RNA, Neoplasm/genetics , S Phase Cell Cycle Checkpoints , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Vesicular Transport Proteins/genetics
7.
Anal Biochem ; 596: 113636, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32081619

ABSTRACT

A procedure is described to measure curcumin (C), demethoxycurcumin (DMC), bisdemethoxycurcumin (BDMC), tetrahydrocurcumim (TC) and their glucuronidated metabolites (CG, DMCG, and BDMCG) in plasma, brain, liver and tumor samples. The procedure involves converting the analytes to their boron difluoride derivatives and analyzing them by combined liquid chromatography coupled to an ion trap mass spectrometer operating in the negative ion MSn scan mode. The method has superb limits of detection of 0.01 nM for all curcuminoids and 0.5 nM for TC and the glucuroniated metabolites, and several representative chromatograms of biological samples containing these analytes are provided. In addition, the pharmacokinetic profile of these compounds in one human who daily consumed an over-the-counter curcuminoid product shows the peak and changes in circulating concentrations achieved by this mode of administration.


Subject(s)
Boranes/chemistry , Diarylheptanoids/blood , Animals , Chromatography, Liquid , Diarylheptanoids/chemistry , Diarylheptanoids/isolation & purification , Healthy Volunteers , Humans , Mass Spectrometry , Mice , Molecular Structure
8.
Cancer ; 126(8): 1668-1682, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32022261

ABSTRACT

BACKGROUND: Although curcumin's effect on head and neck cancer has been studied in vitro and in vivo, to the authors' knowledge its efficacy is limited by poor systemic absorption from oral administration. APG-157 is a botanical drug containing multiple polyphenols, including curcumin, developed under the US Food and Drug Administration's Botanical Drug Development, that delivers the active components to oromucosal tissues near the tumor target. METHODS: A double-blind, randomized, placebo-controlled, phase 1 clinical trial was conducted with APG-157 in 13 normal subjects and 12 patients with oral cancer. Two doses, 100 mg or 200 mg, were delivered transorally every hour for 3 hours. Blood and saliva were collected before and 1 hour, 2 hours, 3 hours, and 24 hours after treatment. Electrocardiograms and blood tests did not demonstrate any toxicity. RESULTS: Treatment with APG-157 resulted in circulating concentrations of curcumin and analogs peaking at 3 hours with reduced IL-1ß, IL-6, and IL-8 concentrations in the salivary supernatant fluid of patients with cancer. Salivary microbial flora analysis showed a reduction in Bacteroidetes species in cancer subjects. RNA and immunofluorescence analyses of tumor tissues of a subject demonstrated increased expression of genes associated with differentiation and T-cell recruitment to the tumor microenvironment. CONCLUSIONS: The results of the current study suggested that APG-157 could serve as a therapeutic drug in combination with immunotherapy. LAY SUMMARY: Curcumin has been shown to suppress tumor cells because of its antioxidant and anti-inflammatory properties. However, its effectiveness has been limited by poor absorption when delivered orally. Subjects with oral cancer were given oral APG-157, a botanical drug containing multiple polyphenols, including curcumin. Curcumin was found in the blood and in tumor tissues. Inflammatory markers and Bacteroides species were found to be decreased in the saliva, and immune T cells were increased in the tumor tissue. APG-157 is absorbed well, reduces inflammation, and attracts T cells to the tumor, suggesting its potential use in combination with immunotherapy drugs.


Subject(s)
Absorption, Physiological/drug effects , Antineoplastic Agents/therapeutic use , Cytokines/antagonists & inhibitors , Microbiota/drug effects , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Adult , Aged , Curcumin/therapeutic use , Cytokines/metabolism , Double-Blind Method , Female , Humans , Inflammation/metabolism , Male , Middle Aged , Polyphenols/therapeutic use , Saliva/microbiology , Tumor Microenvironment/drug effects
9.
Mol Cell Biol ; 36(12): 1776-92, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27090639

ABSTRACT

We and others have shown that the cystatin E/M gene is inactivated in primary human tumors, pointing to its role as a tumor suppressor gene. However, the molecular mechanism of tumor suppression is not yet understood. Using plasmid-directed cystatin E/M gene overexpression, a lentivirus-mediated tetracycline-inducible vector system, and human papillomavirus 16 (HPV 16) E6 and E7 gene-immortalized normal human epidermal keratinocytes, we demonstrated intracellular and non-cell-autonomous apoptotic growth inhibition of tumor cell lines and that growth inhibition is associated with cytoplasmic retention of NF-κB. We further demonstrated decreased phosphorylation of IκB kinase (IKKß) and IκBα in the presence of tumor necrosis factor alpha (TNF-α), confirming the role of cystatin E/M in the regulation of the NF-κB signaling pathway. Growth suppression of nude mouse xenograft tumors carrying a tetracycline-inducible vector system was observed with the addition of doxycycline in drinking water, confirming that the cystatin E/M gene is a tumor suppressor gene. Finally, immunohistochemical analyses of cervical carcinoma in situ and primary tumors have shown a statistically significant inverse relationship between the expression of cystatin E/M and cathepsin L and a direct relationship between the loss of cystatin E/M expression and nuclear expression of NF-κB. We therefore propose that the cystatin E/M suppressor gene plays an important role in the regulation of NF-κB.


Subject(s)
Cystatin M/metabolism , Cytoplasm/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uterine Cervical Neoplasms/pathology , Animals , Cathepsin L/metabolism , Cell Line, Tumor , Cell Proliferation , Cystatin M/genetics , Doxycycline/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Genetic Vectors/pharmacology , HeLa Cells , Humans , Lentivirus/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
10.
Oncotarget ; 6(21): 18504-17, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26098778

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer, with 600,000 new cases every year worldwide. Although chemotherapeutics exist, five-year survival is only 50%. New strategies to overcome drug resistance are required to improve HNSCC treatment. Curcumin-difluorinated (CDF), a synthetic analog of curcumin, was packaged in liposomes and used to evaluate growth inhibition of cisplatin resistant HNSCC cell lines CCL-23R and UM-SCC-1R generated from the parental cell lines CCL-23 and UM-SCC-1 respectively. Growth inhibition in vitro and expression levels of the CD44 (cancer stem cell marker), cytokines, and growth factors were investigated after liposomal CDF treatment. The in vivo growth inhibitory effect of liposomal CDF was evaluated in the nude mice xenograft tumor model of UM-SCC-1R and the inhibition of CD44 was measured. Treatment of the resistant cell lines in vitro with liposomal CDF resulted in a statistically significant growth inhibition (p < 0.05). The nude mice xenograft study showed a statistically significant tumor growth inhibition of UM-SCC-1R cells and a reduction in the expression of CD44 (p < 0.05), indicating an inhibitory effect of liposomal CDF on CSCs. Our results demonstrate that delivery of CDF through liposomes may be an effective method for the treatment of cisplatin resistant HNSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Curcumin/pharmacology , Head and Neck Neoplasms/drug therapy , Hydrocarbons, Fluorinated/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Curcumin/analogs & derivatives , Cytokines/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Hyaluronan Receptors/genetics , Liposomes , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 289(50): 34921-37, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25331947

ABSTRACT

The molecular mechanism of p16-mediated senescence in cisplatin-treated cancer cells is not fully understood. Here we show that cisplatin treatment of head and neck cancer cells results in nuclear transport of p16 leading to a molecular modification of NFκB. Chromatin immunoprecipitation assays show that this modification is associated with the inhibition of NFκB interacting with its DNA binding sequences, leading to decreased expression of NFκB-transcribed proteins. LCMS proteomic analysis of LAP-TAP-purified proteins from HeLa cells containing a tetracycline-inducible GFP-S peptide-NFκB expression system identified gigaxonin, an ubiquitin E3 ligase adaptor, as an NFκB-interacting protein. Immunoblotting and siRNA studies confirmed the NFκB-gigaxonin interaction and the dependence of this binding on p16-NFκB binding. Using gel shift assays, we have confirmed p16-NFκB and gigaxonin-NFκB interactions. Furthermore, we have observed increased NFκB ubiquitination with cisplatin treatment that is abolished in the absence of p16 and gigaxonin expression. Analysis of 103 primary tumors has shown that increased nuclear p16 expression correlates with enhanced survival of head and neck cancer patients (p < 0.0000542), indicating the importance of nuclear p16 expression in prognosis. Finally, p16 expression is associated with reduced cytokine expression and the presence of human papilloma virus in chemoradiation-sensitive basaloid tumors. However, the absence of p16 expression is associated with enhanced cytokine expression and the absence of human papilloma virus in aggressive tumors. These results clearly demonstrate that nuclear p16 and gigaxonin play an important role in chemosensitivity of head and neck cancers through ubiquitination of NFκB.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytoskeletal Proteins/metabolism , NF-kappa B/metabolism , Ubiquitination/drug effects , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Human papillomavirus 16/physiology , Humans , Prognosis
13.
PLoS One ; 8(9): e73195, 2013.
Article in English | MEDLINE | ID: mdl-24019906

ABSTRACT

Cellular heterogeneity is an integral part of cancer development and progression. Progression can be associated with emergence of cells that exhibit high phenotypic plasticity (including "de-differentiation" to primitive developmental states), and aggressive behavioral properties (including high tumorigenic potentials). We observed that many biomarkers that are used to identify Cancer Stem Cells (CSC) can label cell subsets in an advanced clinical stage of lung cancer (malignant pleural effusions, or MPE). Thus, CSC-biomarkers may be useful for live sorting functionally distinct cell subsets from individual tumors, which may enable investigators to hone in on the molecular basis for functional heterogeneity. We demonstrate that the CD44(hi) (CD44-high) cancer cell subsets display higher clonal, colony forming potential than CD44(lo) cells (n=3) and are also tumorigenic (n=2/2) when transplanted in mouse xenograft model. The CD44(hi) subsets express different levels of embryonal (de-differentiation) markers or chromatin regulators. In archived lung cancer tissues, ALDH markers co-localize more with CD44 in squamous cell carcinoma (n=5/7) than Adeno Carcinoma (n=1/12). MPE cancer cells and a lung cancer cell line (NCI-H-2122) exhibit chromosomal abnormalities and 1p36 deletion (n=3/3). Since miR-34a maps to the 1p36 deletion site, low miR-34a expression levels were detected in these cells. The colony forming efficiency of CD44(hi) cells, characteristic property of CSC, can be inhibited by mir-34a replacement in these samples. In addition the highly tumorigenic CD44(hi) cells are enriched for cells in the G2 phase of cell cycle.


Subject(s)
Adenocarcinoma/immunology , Biomarkers, Tumor/immunology , Carcinoma, Squamous Cell/immunology , Hyaluronan Receptors/immunology , Lung Neoplasms/immunology , MicroRNAs/genetics , Adenocarcinoma/genetics , Base Sequence , Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 1 , DNA Primers , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Lung Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Cancer Metastasis Rev ; 31(3-4): 733-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22752409

ABSTRACT

Embryonic stem cells divide continuously and differentiate into organs through the expression of specific transcription factors at specific time periods. Differentiated adult stem cells on the other hand remain in quiescent state and divide by receiving cues from the environment (extracellular matrix or niche), as in the case of wound healing from tissue injury or inflammation. Similarly, it is believed that cancer stem cells (CSCs), forming a smaller fraction of the tumor bulk, also remain in a quiescent state. These cells are capable of initiating and propagating neoplastic growth upon receiving environmental cues, such as overexpression of growth factors, cytokines, and chemokines. Candidate CSCs express distinct biomarkers that can be utilized for their identification and isolation. This review focuses on the known and candidate cancer stem cell markers identified in various solid tumors and the promising future of disease management and therapy targeted at these markers. The review also provides details on the differential expression of microRNAs (miRNAs), and the miRNA- and natural product-based therapies that could be applied for the treatment of cancer stem cells.


Subject(s)
Biological Products/therapeutic use , MicroRNAs/physiology , Neoplastic Stem Cells/physiology , AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/analysis , Basigin/analysis , Carrier Proteins/analysis , Glycoproteins/analysis , Humans , Hyaluronan Receptors/analysis , Isoenzymes/analysis , Membrane Proteins/analysis , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/drug effects , Peptides/analysis , Polycomb Repressive Complex 1/physiology , Proto-Oncogene Proteins/physiology , Reactive Oxygen Species/metabolism , Retinal Dehydrogenase/analysis , Stem Cells/physiology , Thyroid Hormones/analysis , Thyroid Hormone-Binding Proteins
15.
Clin Cancer Res ; 17(18): 5953-61, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21821700

ABSTRACT

PURPOSE: To determine whether curcumin would inhibit IκB kinase ß (IKKß) kinase activity and suppress expression of proinflammatory cytokines in head and neck squamous cell carcinoma cancer (HNSCC) patients. EXPERIMENTAL DESIGN: Saliva was collected before and after subjects chewed curcumin tablets. Protein was extracted and IKKß kinase activity measured. Interleukin (IL)-6 and IL-8 levels in the salivary supernatants were measured by ELISA. IL-6, IL-8, and other interleukin were also measured independently with ELISA to confirm the inhibitory effect of curcumin on expression and secretion of salivary cytokines. RESULTS: Curcumin treatment led to a reduction in IKKß kinase activity in the salivary cells of HNSCC patients (P < 0.05). Treatment of UM-SCC1 cells with curcumin as well as with post-curcumin salivary supernatant showed a reduction of IKKß kinase activity. Significant reduction of IL-8 levels (P < 0.05) was seen in post-curcumin samples from patients with dental caries. Although there was reduced IL-8 expression in 8 of 21 post-curcumin samples of HNSCC patients, the data did not reach statistical significance. Saliva samples from HNSCC patients were also analyzed in a blinded fashion for expression of cytokines. IL-10, IFN-γ, IL-12p70, and IL-2 clustered together, and granulocyte macrophage colony stimulating factor and TNF-α clustered together. Log10 ratio analysis showed decrease in expression of all nine cytokines in both the salivary supernatant and salivary cells of curcumin-treated samples. CONCLUSIONS: Curcumin inhibited IKKß kinase activity in the saliva of HNSCC patients, and this inhibition correlated with reduced expression of a number of cytokines. IKKß kinase could be a useful biomarker for detecting the effect of curcumin in head and neck cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/enzymology , Curcumin/pharmacology , Head and Neck Neoplasms/enzymology , I-kappa B Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Salivary Glands/drug effects , Salivary Glands/enzymology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Curcumin/therapeutic use , Cytokines/biosynthesis , Enzyme Activation/drug effects , Head and Neck Neoplasms/drug therapy , Humans , Male , Middle Aged , Pilot Projects , Protein Kinase Inhibitors/therapeutic use , Saliva/drug effects , Saliva/metabolism , Squamous Cell Carcinoma of Head and Neck
16.
J Stem Cells ; 6(3): 155-79, 2011.
Article in English | MEDLINE | ID: mdl-23264999

ABSTRACT

The recent scientific development using stem or other differentiated cells has generated great hopes for treatment of various diseases. Major thrust has been given to formulate country specific laws and regulations considering international guidelines to conduct research and clinical applications of "Cell Based Therapy" (CBT) all over the world. Attempts have made in this review to discuss the current policies that are practiced by various countries in the areas related to CBT with special emphasis on CBT related research and development in India. The two major funding agencies of Government of India e.g. Department of Biotechnology (DBT) and Indian Council of Medical Research (ICMR), have jointly formulated the "Guidelines for Stem Cell Research and Therapy" in 2007 which requires update and revision. Based on the review of the current world scenario of CBT research and development, suggestions have been made for the development of a new CBT policy that will help in progress of research and patient treatment in India.


Subject(s)
Biomedical Research/legislation & jurisprudence , Cell Transplantation/legislation & jurisprudence , Government Regulation , Health Policy/legislation & jurisprudence , Animals , Biomedical Research/economics , Cell Transplantation/adverse effects , Cell Transplantation/economics , Dendritic Cells/transplantation , Endothelial Cells/transplantation , Genetic Therapy/legislation & jurisprudence , Health Policy/economics , Humans , India , Patient Safety/legislation & jurisprudence , Practice Guidelines as Topic , Research Support as Topic/legislation & jurisprudence , Stem Cell Research/legislation & jurisprudence , Stem Cell Transplantation/legislation & jurisprudence
17.
PLoS One ; 4(6): e5884, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19536353

ABSTRACT

Malignant Pleural Effusions (MPE) may be useful as a model to study hierarchical progression of cancer and/or intratumoral heterogeneity. To strengthen the rationale for developing the MPE-model for these purposes, we set out to find evidence for the presence of cancer stem cells (CSC) in MPE and demonstrate an ability to sustain intratumoral heterogeneity in MPE-primary cultures. Our studies show that candidate lung CSC-expression signatures (PTEN, OCT4, hTERT, Bmi1, EZH2 and SUZ12) are evident in cell pellets isolated from MPE, and MPE-cytopathology also labels candidate-CSC (CD44, cMET, MDR-1, ALDH) subpopulations. Moreover, in primary cultures that use MPE as the source of both tumor cells and the tumor microenvironment (TME), candidate CSC are maintained over time. This allows us to live-sort candidate CSC-fractions from the MPE-tumor mix on the basis of surface markers (CD44, c-MET, uPAR, MDR-1) or differences in xenobiotic metabolism (ALDH). Thus, MPE-primary cultures provide an avenue to extract candidate CSC populations from individual (isogenic) MPE-tumors. This will allow us to test whether these cells can be discriminated in functional bioassays. Tumor heterogeneity in MPE-primary cultures is evidenced by variable immunolabeling, differences in colony-morphology, and differences in proliferation rates of cell subpopulations. Collectively, these data justify the ongoing development of the MPE-model for the investigation of intratumoral heterogeneity, tumor-TME interactions, and phenotypic validation of candidate lung CSC, in addition to providing direction for the pre-clinical development of rational therapeutics.


Subject(s)
Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Pleural Effusion, Malignant/pathology , Aldehyde Dehydrogenase/metabolism , Cell Line, Tumor , Cell Separation , Female , Flow Cytometry , Humans , Male , Neoplastic Stem Cells/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Smoking/adverse effects , Stem Cells/cytology
18.
J Immunol Methods ; 312(1-2): 94-104, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16626731

ABSTRACT

The level of antigen loading can impact on the capacity for dendritic cells (DC) to activate T cell responses. Several different approaches to adenoviral (Ad)-based transduction were therefore assessed for their effect on both transgene expression and T cell activation. While a conventional E1(-)/E3Delta Ad vector (Ad/GFP) produced a concentration-dependent expression of GFP, a modified vector expressing Arginine-Glycine-Aspartic Acid (RGD) sequence on its fiber knob (Ad-RGD/GFP) enhanced transgene expression by 9-20-fold at each MOI. The addition of centrifugal force (2000xg) during DC transduction with Ad/GFP also increased expression up to 20-fold. However, combining centrifugation with the Ad-RGD/GFP vector produced no effect on transduction rate and only a 1.5- to 2-fold increase in GFP expression, suggesting overlapping mechanisms of action. Consistent with this, exogenous RGD peptide blocked transduction regardless of the vector used, or the addition of centrifugal force, and transduction was primarily limited to DC expressing the CD51 integrin receptor. Ad vectors expressing ovalbumin (OVA) were used to assess transduced DC for their capacity to activate OVA-specific T cells. We observed a significant relationship between transgene expression and the capacity for T cell activation regardless of whether transgene expression was increased by using a higher MOI, an RGD-modified vector, or by employing centrifugal force. Furthermore, combining these approaches produced synergistic effects on T cell activation. We conclude that RGD-modified vectors and centrifugation both enhance DC transduction by increasing entry via integrin receptors and that the capacity for T cell activation can be optimized by combining approaches to achieve the highest possible level of transgene expression.


Subject(s)
Dendritic Cells/immunology , Genetic Vectors/genetics , Oligopeptides/genetics , Transduction, Genetic/methods , Adenoviridae/genetics , Animals , Antigens, Differentiation/analysis , Centrifugation , Coculture Techniques , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Integrin alpha Chains/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transgenes
19.
J Leukoc Biol ; 79(6): 1271-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16574766

ABSTRACT

Dendritic cells (DC) play a central role in antigen presentation and are often targeted by adenoviral (Ad)-based gene therapy. However, DC lack the coxsackie-Ad receptor, and little is known about the process by which they acquire and present Ad-encoded antigens. We examined the expression of alpha(v)beta3 integrins (CD51/CD61) on mouse bone marrow-derived DC (BM-DC) and their susceptibility to transduction by Ad vectors. Less than 10% of BM-DC precursors expressed CD51, but expression increased over time in culture with granulocyte macrophage-colony stimulating factor (GM-CSF)/interleukin (IL)-4. After 7 days, 28 +/- 1.7% of CD11c+ DC expressed high levels of CD51 (CD51(hi)), and the remaining DC expressed low levels of CD51 (CD51(lo)). CD51(hi) CD express higher major histocompatibility complex type 1 (MHC I); however, both of the DC subsets expressed similar levels of MHC II and costimulatory molecules. When exposed to a first-generation Ad vector, transgene expression was restricted to the CD51(hi) DC subset and blocked by soluble peptides expressing an arginine, glycine, aspartic acid (RGD) sequence, confirming the role of integrins in viral entry. Consistent with this, a modified Ad expressing an RGD-binding sequence in its fiber knob (Ad-RGD) transduced the CD51(hi) DC subset with significantly higher efficiency. When BM-DC were transduced with an Ad-expressing ovalbumin (Ad-OVA), the CD51(hi) subset proved superior in activating OT-I (T cell receptor-OVA) T cells. Similar to in vitro effects, systemic administration of GM-CSF/IL-4 increased the expression of CD51 on splenic DC and rendered these cells susceptible to Ad transduction. These results suggest that a limited subset of DC expressing high levels of alpha(v)beta3 integrins is preferentially transduced by Ad vectors and activates CD8+ T cell responses against Ad-encoded antigens.


Subject(s)
Adenoviridae/immunology , Antigen Presentation , Antigens, Viral/immunology , Dendritic Cells/immunology , Genetic Vectors/immunology , Integrin alphaVbeta3/analysis , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Bone Marrow Cells/chemistry , Bone Marrow Cells/classification , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Dendritic Cells/chemistry , Dendritic Cells/classification , Dendritic Cells/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , H-2 Antigens/immunology , Integrin alphaV/biosynthesis , Integrin alphaV/genetics , Integrin beta3/biosynthesis , Integrin beta3/genetics , Interleukin-4/pharmacology , Male , Mice , Mice, Inbred C57BL , Oligopeptides/pharmacology , Ovalbumin/immunology , Recombinant Proteins/pharmacology , Spleen/cytology , Transduction, Genetic , Transgenes
20.
Viral Immunol ; 17(2): 182-96, 2004.
Article in English | MEDLINE | ID: mdl-15279698

ABSTRACT

The past decade has produced significant advances in our understanding of antigen-presenting cells, tumor antigens, and other components of the immune response to cancer. Gene-based vaccination is emerging as one of the more promising approaches for loading dendritic cells (DC) with tumor-associated antigens. In this respect, it is proposed that adenoviral (AdV) vectors can deliver high antigen concentrations, promote effective processing and MHC expression, and stimulate potent cell-mediated immunity. While AdV vectors have performed well in pre-clinical vaccine models, their application to patient care has limitations. The in vivo administration of AdV vectors is associated with both innate and adaptive host responses that result in tissue inflammation and injury, viral neutralization, and premature clearance of AdV-transduced cells. A variety of strategies have been developed to address these limitations. The ideal vaccine would avoid vector-related immune responses, have relative specificity for transducing DC, and induce high levels of transgene expression. This review describes the range of host responses to AdV vaccines, identifies strategies to reduce viral recognition and enhance transgene antigen expression, and suggests future approaches to vector development and administration. There is every reason to believe that safer and more effective forms of AdV-based vaccines can be developed and applied to patient therapy.


Subject(s)
Adenoviruses, Human/immunology , Cancer Vaccines/therapeutic use , Neoplasms/therapy , Adenoviruses, Human/genetics , Animals , Genetic Therapy/methods , Genetic Therapy/trends , Genetic Vectors/immunology , Humans , Immunotherapy/methods , Immunotherapy/trends , Neoplasms/genetics , Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...