Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 1700, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402224

ABSTRACT

The Ataxia telangiectasia and Rad3-related (ATR) inhibitor ceralasertib in combination with the PD-L1 antibody durvalumab demonstrated encouraging clinical benefit in melanoma and lung cancer patients who progressed on immunotherapy. Here we show that modelling of intermittent ceralasertib treatment in mouse tumor models reveals CD8+ T-cell dependent antitumor activity, which is separate from the effects on tumor cells. Ceralasertib suppresses proliferating CD8+ T-cells on treatment which is rapidly reversed off-treatment. Ceralasertib causes up-regulation of type I interferon (IFNI) pathway in cancer patients and in tumor-bearing mice. IFNI is experimentally found to be a major mediator of antitumor activity of ceralasertib in combination with PD-L1 antibody. Improvement of T-cell function after ceralasertib treatment is linked to changes in myeloid cells in the tumor microenvironment. IFNI also promotes anti-proliferative effects of ceralasertib on tumor cells. Here, we report that broad immunomodulatory changes following intermittent ATR inhibition underpins the clinical therapeutic benefit and indicates its wider impact on antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Indoles , Morpholines , Neoplasms , Pyrimidines , Sulfonamides , Humans , Animals , Mice , B7-H1 Antigen , Tumor Microenvironment , Cell Line, Tumor , Immunotherapy , Disease Models, Animal , Ataxia Telangiectasia Mutated Proteins
2.
Cancers (Basel) ; 15(16)2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37627223

ABSTRACT

Ataxia-telangiectasia mutated gene (ATM) is a key component of the DNA damage response (DDR) and double-strand break repair pathway. The functional loss of ATM (ATM deficiency) is hypothesised to enhance sensitivity to DDR inhibitors (DDRi). Whole-exome sequencing (WES), immunohistochemistry (IHC), and Western blotting (WB) were used to characterise the baseline ATM status across a panel of ATM mutated patient-derived xenograft (PDX) models from a range of tumour types. Antitumour efficacy was assessed with poly(ADP-ribose)polymerase (PARP, olaparib), ataxia- telangiectasia and rad3-related protein (ATR, AZD6738), and DNA-dependent protein kinase (DNA-PK, AZD7648) inhibitors as a monotherapy or in combination to associate responses with ATM status. Biallelic truncation/frameshift ATM mutations were linked to ATM protein loss while monoallelic or missense mutations, including the clinically relevant recurrent R3008H mutation, did not confer ATM protein loss by IHC. DDRi agents showed a mixed response across the PDX's but with a general trend toward greater activity, particularly in combination in models with biallelic ATM mutation and protein loss. A PDX with an ATM splice-site mutation, 2127T > C, with a high relative baseline ATM expression and KAP1 phosphorylation responded to all DDRi treatments. These data highlight the heterogeneity and complexity in describing targetable ATM-deficiencies and the fact that current patient selection biomarker methods remain imperfect; although, complete ATM loss was best able to enrich for DDRi sensitivity.

3.
Mutagenesis ; 37(5-6): 227-237, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36426854

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer death in the UK. Novel therapeutic prevention strategies to inhibit the development and progression of CRC would be invaluable. Potential contenders include low toxicity agents such as dietary-derived agents or repurposed drugs. However, in vitro and in vivo models used in drug development often do not take into account the heterogeneity of tumours or the tumour microenvironment. This limits translation to a clinical setting. Our objectives were to develop an ex vivo method utilizing CRC and adenoma patient-derived explants (PDEs) which facilitates screening of drugs, assessment of toxicity, and efficacy. Our aims were to use a multiplexed immunofluorescence approach to demonstrate the viability of colorectal tissue PDEs, and the ability to assess immune cell composition and interactions. Using clinically achievable concentrations of curcumin, we show a correlation between curcumin-induced tumour and stromal apoptosis (P < .001) in adenomas and cancers; higher stromal content is associated with poorer outcomes. B cell (CD20+ve) and T cell (CD3+ve) density of immune cells within tumour regions in control samples correlated with curcumin-induced tumour apoptosis (P < .001 and P < .05, respectively), suggesting curcumin-induced apoptosis is potentially predicted by baseline measures of immune cells. A decrease in distance between T cells (CD3+ve) and cytokeratin+ve cells was observed, indicating movement of T cells (CD3+ve) towards the tumour margin (P < .001); this change is consistent with an immune environment associated with improved outcomes. Concurrently, an increase in distance between T cells (CD3+ve) and B cells (CD20+ve) was detected following curcumin treatment (P < .001), which may result in a less immunosuppressive tumour milieu. The colorectal tissue PDE model offers significant potential for simultaneously assessing multiple biomarkers in response to drug exposure allowing a greater understanding of mechanisms of action and efficacy in relevant target tissues, that maintain both their structural integrity and immune cell compartments.


Subject(s)
Adenoma , Colorectal Neoplasms , Humans , Adenoma/drug therapy , Colorectal Neoplasms/drug therapy , Tumor Microenvironment
4.
J Med Chem ; 64(18): 13524-13539, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34478292

ABSTRACT

Inhibition of Mer and Axl kinases has been implicated as a potential way to improve the efficacy of current immuno-oncology therapeutics by restoring the innate immune response in the tumor microenvironment. Highly selective dual Mer/Axl kinase inhibitors are required to validate this hypothesis. Starting from hits from a DNA-encoded library screen, we optimized an imidazo[1,2-a]pyridine series using structure-based compound design to improve potency and reduce lipophilicity, resulting in a highly selective in vivo probe compound 32. We demonstrated dose-dependent in vivo efficacy and target engagement in Mer- and Axl-dependent efficacy models using two structurally differentiated and selective dual Mer/Axl inhibitors. Additionally, in vivo efficacy was observed in a preclinical MC38 immuno-oncology model in combination with anti-PD1 antibodies and ionizing radiation.


Subject(s)
Antineoplastic Agents/therapeutic use , Imidazoles/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Imidazoles/chemical synthesis , Male , Mice, Inbred C57BL , Mice, Nude , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins/metabolism , Pyridines/chemical synthesis , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , c-Mer Tyrosine Kinase/metabolism , Axl Receptor Tyrosine Kinase
5.
Clin Cancer Res ; 27(15): 4353-4366, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34011558

ABSTRACT

PURPOSE: Combining radiotherapy (RT) with DNA damage response inhibitors may lead to increased tumor cell death through radiosensitization. DNA-dependent protein kinase (DNA-PK) plays an important role in DNA double-strand break repair via the nonhomologous end joining (NHEJ) pathway. We hypothesized that in addition to a radiosensitizing effect from the combination of RT with AZD7648, a potent and specific inhibitor of DNA-PK, combination therapy may also lead to modulation of an anticancer immune response. EXPERIMENTAL DESIGN: AZD7648 and RT efficacy, as monotherapy and in combination, was investigated in fully immunocompetent mice in MC38, CT26, and B16-F10 models. Immunologic consequences were analyzed by gene expression and flow-cytometric analysis. RESULTS: AZD7648, when delivered in combination with RT, induced complete tumor regressions in a significant proportion of mice. The antitumor efficacy was dependent on the presence of CD8+ T cells but independent of NK cells. Analysis of the tumor microenvironment revealed a reduction in T-cell PD-1 expression, increased NK-cell granzyme B expression, and elevated type I IFN signaling in mice treated with the combination when compared with RT treatment alone. Blocking of the type I IFN receptor in vivo also demonstrated a critical role for type I IFN in tumor growth control following combined therapy. Finally, this combination was able to generate tumor antigen-specific immunologic memory capable of suppressing tumor growth following rechallenge. CONCLUSIONS: Blocking the NHEJ DNA repair pathway with AZD7648 in combination with RT leads to durable immune-mediated tumor control.


Subject(s)
Cell Line, Tumor/radiation effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Interferon Type I/drug effects , Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrans/pharmacology , Radiation-Sensitizing Agents/pharmacology , Triazoles/pharmacology , Animals , Mice
6.
Nat Commun ; 10(1): 5065, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31699977

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a critical player in the DNA damage response (DDR) and instrumental in the non-homologous end-joining pathway (NHEJ) used to detect and repair DNA double-strand breaks (DSBs). We demonstrate that the potent and highly selective DNA-PK inhibitor, AZD7648, is an efficient sensitizer of radiation- and doxorubicin-induced DNA damage, with combinations in xenograft and patient-derived xenograft (PDX) models inducing sustained regressions. Using ATM-deficient cells, we demonstrate that AZD7648, in combination with the PARP inhibitor olaparib, increases genomic instability, resulting in cell growth inhibition and apoptosis. AZD7648 enhanced olaparib efficacy across a range of doses and schedules in xenograft and PDX models, enabling sustained tumour regression and providing a clear rationale for its clinical investigation. Through its differentiated mechanism of action as an NHEJ inhibitor, AZD7648 complements the current armamentarium of DDR-targeted agents and has potential in combination with these agents to achieve deeper responses to current therapies.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Drug Synergism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrans/pharmacology , Radiation Tolerance/drug effects , Triazoles/pharmacology , A549 Cells , Animals , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Genomic Instability/drug effects , Humans , Lung Neoplasms , Mice , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Polyethylene Glycols/pharmacology , Radiotherapy , Xenograft Model Antitumor Assays
7.
Sci Rep ; 8(1): 12096, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108309

ABSTRACT

Tumors deficient in the urea cycle enzymes argininosuccinate synthase-1 (ASS1) and ornithine transcarbamylase (OTC) are unable to synthesize arginine and can be targeted using arginine-deprivation therapy. Here, we show that colorectal cancers (CRCs) display negligible expression of OTC and, in subset of cases, ASS1 proteins. CRC cells fail to grow in arginine-free medium and dietary arginine deprivation slows growth of cancer cells implanted into immunocompromised mice. Moreover, we report that clinically-formulated arginine-degrading enzymes are effective anticancer drugs in CRC. Pegylated arginine deiminase (ADI-PEG20), which degrades arginine to citrulline and ammonia, affects growth of ASS1-negative cells, whereas recombinant human arginase-1 (rhArg1peg5000), which degrades arginine into urea and ornithine, is effective against a broad spectrum of OTC-negative CRC cell lines. This reflects the inability of CRC cells to recycle citrulline and ornithine into the urea cycle. Finally, we show that arginase antagonizes chemotherapeutic drugs oxaliplatin and 5-fluorouracil (5-FU), whereas ADI-PEG20 synergizes with oxaliplatin in ASS1-negative cell lines and appears to interact with 5-fluorouracil independently of ASS1 status. Overall, we conclude that CRC is amenable to arginine-deprivation therapy, but we warrant caution when combining arginine deprivation with standard chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arginine/antagonists & inhibitors , Argininosuccinate Synthase/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arginase/pharmacology , Arginase/therapeutic use , Arginine/metabolism , Cell Line, Tumor , Colon/pathology , Colorectal Neoplasms/mortality , Drug Interactions , Drug Synergism , Feasibility Studies , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Hydrolases/pharmacology , Hydrolases/therapeutic use , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Male , Mice , Ornithine Carbamoyltransferase/metabolism , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Retrospective Studies , Treatment Outcome , Urea/metabolism , Xenograft Model Antitumor Assays
8.
Mol Nutr Food Res ; 60(6): 1295-309, 2016 06.
Article in English | MEDLINE | ID: mdl-27060283

ABSTRACT

Diet has been linked to an overwhelming proportion of cancers. Current chemotherapy and targeted therapies are limited by toxicity and the development of resistance against these treatments results in cancer recurrence or progression. In vitro evidence indicates that a number of dietary-derived agents have activity against a highly tumorigenic, chemoradiotherapy resistant population of cells within a tumour. This population is associated with cancer recurrence and is therefore clinically significant. Targeting this subpopulation, termed cancer stem-like cells with dietary-derived agents provides a potentially low toxicity strategy to enhance current treatment regimens. In addition, dietary-derived compounds also provide a novel approach to cancer prevention strategies. This review focusses on selected diet-derived agents that have been shown to specifically target cancer stem-like cells using in vivo models, or in clinical trials. Furthermore, the potential limitations of these studies are discussed, and areas of research that need to be addressed to allow successful translation of dietary-derived agents to the clinical arena are highlighted.


Subject(s)
Diet , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Curcumin/pharmacology , Disease Models, Animal , Flavonoids/pharmacology , Humans , Isothiocyanates/pharmacology , Randomized Controlled Trials as Topic , Resveratrol , Stilbenes/pharmacology , Sulfoxides , Vitamin A/pharmacology
9.
Food Chem Toxicol ; 92: 8-16, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26995228

ABSTRACT

Colon cancer stem cells (CSCs) offer a novel paradigm for colorectal cancer (CRC) treatment and dietary polyphenols may contribute to battle these cells. Specifically, polyphenol-derived colon metabolites have the potential to interact with and affect colon CSCs. We herein report the effects against colon CSCs of two mixtures of ellagitannin (ET) metabolites, ellagic acid (EA) and the gut microbiota-derived urolithins (Uro) at concentrations detected in the human colon tissues following the intake of ET-containing products (pomegranate, walnuts). These mixtures reduce phenotypic and molecular features in two models of colon CSCs: Caco-2 cells and primary tumour cells from a patient with CRC. The mixture containing mostly Uro-A (85% Uro-A, 10% Uro-C, 5% EA) was most effective at inhibiting the number and size of colonospheres and aldehyde dehydrogenase activity (ALDH, a marker of chemoresistance) whereas the mixture containing less Uro-A but IsoUro-A and Uro-B (30% Uro-A, 50% IsoUro-A, 10% Uro-B, 5% Uro-C, 5% EA) had some effects on the number and size of colonospheres but not on ALDH. These data support a role for polyphenols metabolites in the control of colon cancer chemoresistance and relapse and encourage the research on the effects of polyphenols against CSCs.


Subject(s)
Colonic Neoplasms/drug therapy , Coumarins/pharmacology , Ellagic Acid/pharmacology , Hydrolyzable Tannins/metabolism , Neoplastic Stem Cells/drug effects , Biomarkers, Tumor/metabolism , Colonic Neoplasms/pathology , Drug Therapy, Combination , Humans , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured
10.
Sci Transl Med ; 7(298): 298ra117, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26223300

ABSTRACT

Resveratrol is widely promoted as a potential cancer chemopreventive agent, but a lack of information on the optimal dose prohibits rationally designed trials to assess efficacy. To challenge the assumption that "more is better," we compared the pharmacokinetics and activity of a dietary dose with an intake 200 times higher. The dose-response relationship for concentrations generated and the metabolite profile of [(14)C]-resveratrol in colorectal tissue of cancer patients helped us to define clinically achievable levels. In Apc(Min) mice (a model of colorectal carcinogenesis) that received a high-fat diet, the low resveratrol dose suppressed intestinal adenoma development more potently than did the higher dose. Efficacy correlated with activation of adenosine monophosphate-activated protein kinase (AMPK) and increased expression of the senescence marker p21. Nonlinear dose responses were observed for AMPK and mechanistic target of rapamycin (mTOR) signaling in mouse adenoma cells, culminating in autophagy and senescence. In human colorectal tissues exposed to low dietary concentrations of resveratrol ex vivo, we measured enhanced AMPK phosphorylation and autophagy. The expression of the cytoprotective NAD(P)H dehydrogenase, quinone 1 (NQO1) enzyme was also increased in tissues from cancer patients participating in our [(14)C]-resveratrol trial. These findings warrant a revision of developmental strategies for diet-derived agents designed to achieve cancer chemoprevention.


Subject(s)
Adenoma/drug therapy , Antineoplastic Agents, Phytogenic/administration & dosage , Colorectal Neoplasms/drug therapy , Stilbenes/administration & dosage , AMP-Activated Protein Kinases/metabolism , Adenoma/metabolism , Adenoma/pathology , Animals , Autophagy/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Diet, High-Fat , Dose-Response Relationship, Drug , Humans , Mice , Resveratrol , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
11.
Cancer Lett ; 364(2): 135-41, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-25979230

ABSTRACT

In vitro and pre-clinical studies have suggested that addition of the diet-derived agent curcumin may provide a suitable adjunct to enhance efficacy of chemotherapy in models of colorectal cancer. However, the majority of evidence for this currently derives from established cell lines. Here, we utilised patient-derived colorectal liver metastases (CRLM) to assess whether curcumin may provide added benefit over 5-fluorouracil (5-FU) and oxaliplatin (FOLFOX) in cancer stem cell (CSC) models. Combination of curcumin with FOLFOX chemotherapy was then assessed clinically in a phase I dose escalation study. Curcumin alone and in combination significantly reduced spheroid number in CRLM CSC models, and decreased the number of cells with high aldehyde dehydrogenase activity (ALDH(high)/CD133(-)). Addition of curcumin to oxaliplatin/5-FU enhanced anti-proliferative and pro-apoptotic effects in a proportion of patient-derived explants, whilst reducing expression of stem cell-associated markers ALDH and CD133. The phase I dose escalation study revealed curcumin to be a safe and tolerable adjunct to FOLFOX chemotherapy in patients with CRLM (n = 12) at doses up to 2 grams daily. Curcumin may provide added benefit in subsets of patients when administered with FOLFOX, and is a well-tolerated chemotherapy adjunct.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Curcumin/administration & dosage , Curcumin/adverse effects , Dose-Response Relationship, Drug , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Heterografts , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Liver Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Spheroids, Cellular
12.
PLoS One ; 10(2): e0117776, 2015.
Article in English | MEDLINE | ID: mdl-25658706

ABSTRACT

Tumor initiating cells (TIC) are increasingly being put forward as a potential target for intervention within colorectal cancer. Whilst characterisation and outgrowth of these cells has been extensively undertaken in primary colorectal cancers, few data are available describing characteristics within the metastatic setting. Tissue was obtained from patients undergoing surgical resection for colorectal liver metastases, and processed into single cell suspension for assessment. Tumor initiating cells from liver metastases were characterised using combinations of EPCAM, Aldehyde dehydrogenase activity, CD133 and CD26. CD133 expression was significantly lower in patients who had received chemotherapy, but this was accounted for by a decrease observed in the male patient cohort only. ALDHhigh populations were rare (0.4 and 0.3% for EPCAM+/ALDHhigh/CD133- and EPCAM+/ALDHhigh/CD133+ populations respectively) and below the limits of detection in 28% of samples. Spheroid outgrowth of metastatic tumor cells across all samples could not be readily achieved using standard spheroid-formation techniques, thus requiring further method validation to reliably propagate cells from the majority of tissues. Spheroid formation was not enhanced using additional growth factors or fibroblast co-culture, but once cells were passaged through NOD-SCID mice, spheroid formation was observed in 82% samples, accompanied by a significant increase in CD26. Order of spheroid forming ability was ALDHhigh>CD133>CD26. Samples sorted by these markers each had the ability to reform ALDHhigh, CD133 and CD26 positive populations to a similar extent, suggestive of a high degree of plasticity for each population. Ex vivo TIC models are increasingly being utilised to assess efficacy of therapeutic interventions. It is therefore essential that such investigations use well-characterised models that are able to sustain TIC populations across a large patient cohort in order that the inherent heterogeneity observed in cancer populations is maintained.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , AC133 Antigen , Aged , Aged, 80 and over , Aldehyde Dehydrogenase/metabolism , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Coculture Techniques , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dipeptidyl Peptidase 4/metabolism , Epithelial Cell Adhesion Molecule , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Glycoproteins/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Peptides/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Sci Transl Med ; 5(205): 205ra133, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24089405

ABSTRACT

The phytochemical resveratrol has been shown to exert numerous health benefits in preclinical studies, but its rapid metabolism and resulting poor bioavailability may limit translation of these effects to humans. Resveratrol metabolites might contribute to in vivo activity through regeneration of the parent compound. We present quantitation of sulfate and glucuronide conjugates of resveratrol in human plasma and tissue after repeated ingestion of resveratrol by volunteers and cancer patients, respectively. Subsequent pharmacokinetic characterization of a mixture of resveratrol-3-O-sulfate and resveratrol-4'-O-sulfate in mice showed that these metabolites are absorbed orally but have low bioavailabilities of ~14 and 3%, respectively. Sulfate hydrolysis in vivo liberated free resveratrol, which accounted for ~2% of the total resveratrol species present in mouse plasma. Monosulfate metabolites were also converted to the parent in human colorectal cells. The extent of cellular uptake was dependent on specific membrane transporters and dictated antiproliferative activity. Sulfate metabolites induced autophagy and senescence in human cancer cells; these effects were abrogated by inclusion of a sulfatase inhibitor, which reduced intracellular resveratrol. Together, our findings suggest that resveratrol is delivered to target tissues in a stable sulfate-conjugated form and that the parent compound is gradually regenerated in selected cells and may give rise to the beneficial effects in vivo. At doses considered to be safe in humans, resveratrol generated via this route may be of greater importance than the unmetabolized form.


Subject(s)
Autophagy , Cellular Senescence , Intracellular Space/metabolism , Stilbenes/metabolism , Sulfates/metabolism , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation , Cellular Senescence/drug effects , Chromatography, High Pressure Liquid , Colorectal Neoplasms/blood , Glucuronides/blood , Humans , Intracellular Space/drug effects , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Resveratrol , Stilbenes/blood , Stilbenes/pharmacology
14.
Mol Nutr Food Res ; 57(9): 1630-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23900994

ABSTRACT

Many cancers contain cell subpopulations that display characteristics of stem cells. These cells are characterised by their ability to self-renew, form differentiated progeny and develop resistance to chemotherapeutic strategies. Cancer stem cells may utilise many of the same signalling pathways as normal stem cells including Wnt, Notch and Hedgehog. The dietary agent curcumin exerts a plethora of anti-carcinogenic effects both in vitro and in vivo, and can also inhibit many of the signalling pathways associated with stem cell biology. Emerging evidence suggests that curcumin can exert its anti-carcinogenic activity via targeting cancer stem cells through the disruption of stem cell signalling pathways. In this review we summarise the ability of curcumin to interfere with signalling pathways Wnt, Hedgehog, Notch, Signal Transducers and Activator (STAT) and interleukin-8, and report curcumin-induced changes in function and properties of cancer stem cells. We present evidence that the effects of curcumin on cancer stem cells mediate, or contribute to, its anti-carcinogenic activity.


Subject(s)
Anticarcinogenic Agents/pharmacology , Curcumin/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Disease Models, Animal , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Interleukin-8/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism
15.
Mol Nutr Food Res ; 56(5): 775-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22648624

ABSTRACT

SCOPE: Rosmarinic acid (RA), a constituent of culinary herbs is considered to possess cancer chemopreventive properties. It has been shown to inhibit the development of cancer in preclinical models but data are conflicting and whether it can protect against gastrointestinal malignancies in vivo has not been examined. This study aimed to investigate the effect of RA on the development of intestinal adenomas in the Apc(Min) mouse model of colorectal carcinogenesis, and to correlate efficacy with levels of RA achieved in the plasma and gastrointestinal tract. METHODS AND RESULTS: RA inhibited the growth of APC10.1 cells derived from Apc(Min) mouse adenomas, with an IC50 of 43 µM. Consumption of dietary RA (0.3%) by Apc(Min) mice for 8 weeks post weaning decreased adenoma burden by ∼35%, but the difference from controls was not significant. Although RA significantly decreased the frequency of large adenomas, the number of small ones increased. Using a novel validated HPLC assay, average levels of RA in the plasma and intestinal mucosa of these mice were found to be 1.1 µM and 38 nmol/g, respectively. CONCLUSION: Chronic consumption of RA furnished quantifiable levels of parent compound in the plasma and intestinal tract of Apc(Min) mice and may slow adenoma development.


Subject(s)
Adenoma/prevention & control , Anticarcinogenic Agents/pharmacology , Cinnamates/pharmacology , Colorectal Neoplasms/prevention & control , Depsides/pharmacology , Adenoma/genetics , Adenoma/pathology , Animals , Calibration , Chromatography, High Pressure Liquid/methods , Cinnamates/analysis , Cinnamates/pharmacokinetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Depsides/analysis , Depsides/pharmacokinetics , Dietary Supplements , Disease Models, Animal , Drug Screening Assays, Antitumor , Genes, APC , Intestinal Mucosa/drug effects , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestinal Neoplasms/prevention & control , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Reproducibility of Results , Sensitivity and Specificity , Tumor Cells, Cultured , Rosmarinic Acid
16.
Best Pract Res Clin Gastroenterol ; 25(4-5): 519-34, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22122768

ABSTRACT

Curcumin is a naturally occurring phytochemical and an extract of turmeric. Extensive in vitro and in vivo data have paved the way for curcumin to become the subject of clinical trials. Curcumin modulates key signalling pathways important in cellular processes. Numerous mechanisms of action have been elucidated. The potential for clinical efficacy is apparent from benign and malignant disease models. Curcumin has potent anti-inflammatory and anti-neoplastic properties used alone and in combination with standard therapies. Early-phase trials have ascertained pharmacological properties and consistently demonstrate it to be safe and well tolerated. However, bioavailability is limited and efficacious doses have not yet been determined. Evidence of efficacy has been derived from animal models or small clinical trials. There is only finite data supporting the use of curcumin in phase III trials with specific diseases (e.g. ulcerative colitis). However, for the vast majority of conditions additional early-phase studies are required to justify larger trials determining efficacy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Curcumin/therapeutic use , Gastrointestinal Agents/therapeutic use , Gastrointestinal Diseases/drug therapy , Animals , Humans , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL