Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Am J Clin Nutr ; 113(5): 1115-1125, 2021 05 08.
Article in English | MEDLINE | ID: mdl-33675348

ABSTRACT

BACKGROUND: The dietary polyphenol resveratrol prevents various malignancies in preclinical models, including prostate cancer. Despite attempts to translate findings to humans, gaps remain in understanding pharmacokinetic-pharmacodynamic relations and how tissue concentrations affect efficacy. Such information is necessary for dose selection and is particularly important given the low bioavailability of resveratrol. OBJECTIVES: This study aimed to determine concentrations of resveratrol in prostate tissue of men after a dietary-achievable (5 mg) or pharmacological (1 g) dose. We then examined whether clinically relevant concentrations of resveratrol/its metabolites had direct anticancer activity in prostate cell lines. METHODS: A window trial was performed in which patients were allocated to 5 mg or 1 g resveratrol daily, or no intervention, before prostate biopsy. Patients (10/group) ingested resveratrol capsules for 7-14 d before biopsy, with the last dose [14C]-labeled, allowing detection of resveratrol species in prostate tissue using accelerator MS. Cellular uptake and antiproliferative properties of resveratrol/metabolites were assessed in cancer and nonmalignant cell cultures using HPLC with UV detection and cell counting, respectively. RESULTS: [14C]-Resveratrol species were detectable in prostate tissue of all patients analyzed, with mean ± SD concentrations of 0.08 ± 0.04 compared with 22.1 ± 8.2 pmol/mg tissue for the 5 mg and the 1 g dose, respectively. However, total [14C]-resveratrol equivalents in prostate were lower than we previously reported in plasma and colorectum after identical doses. Furthermore, resveratrol was undetectable in prostate tissue; instead, sulfate and glucuronide metabolites dominated. Although resveratrol reduced prostate cell numbers in vitro over 7 d, the concentrations required (≥10 µM) exceeded the plasma maximum concentration. Resveratrol mono-sulfates and glucuronides failed to consistently inhibit cell growth, partly due to poor cellular uptake. CONCLUSIONS: Low tissue concentrations of resveratrol species, coupled with weak antiproliferative activity of its conjugates, suggest daily doses of ≤1 g may not have direct effects on human prostate.This trial was registered at clinicaltrialsregister.eu as EudraCT 2007-002131-91.


Subject(s)
Prostate/metabolism , Resveratrol/metabolism , Resveratrol/pharmacokinetics , Administration, Oral , Antioxidants/administration & dosage , Antioxidants/metabolism , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Carbon Radioisotopes , Cell Line, Tumor , Diet , Dose-Response Relationship, Drug , Drug Administration Routes , Humans , Isotope Labeling , Male , Prostatic Neoplasms/prevention & control , Resveratrol/administration & dosage , Resveratrol/therapeutic use
2.
Ann Transl Med ; 8(5): 172, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32309319

ABSTRACT

BACKGROUND: Pancreatic adenocarcinoma (PAC) is a devastating condition, with the majority of patients presenting with metastatic or locally advanced disease. In these patients their disease is classified as advanced pancreatic cancer (APC), which is incurable and associated with survivals generally of a few months. The overall survival (OS) for pancreatic cancer has not changed significantly in the past forty years with multiple trials demonstrating disappointing results. Immune modulatory cells particularly myeloid derived suppressor cells (MDSCs) and T regulatory cells (Tregs) are important mediators in PAC. Omega 3 fatty acids (ω-3FAs) have been shown to have anti-inflammatory properties and there is now evidence demonstrating the benefit of ω-3FAs in PAC. METHODS: This was a single-center cohort study investigating intravenous ω-3FAs and gemcitabine chemotherapy versus gemcitabine therapy only in patients with APC. Here, we investigated levels of MDSCs and Tregs and examined how these changes correlated with survival. RESULTS: Eighteen trial and nine control patients were recruited. There was a significant benefit in progression-free survival (PFS) in trial compared to control patients (P=0.0003). Median survival in trial patients was 5.65 months compared to 1.8 months in control patients. There was no significant benefit in OS in trial compared to control patients (P=0.13). Median survival in trial patients was 7 months compared to 2.9 months in control patients. MDSCs were significantly decreased in trial patients (P=0.0001) but not control patients. Conversely Tregs were significantly increased in control patients (P=0.005) but not in trial patients. CONCLUSIONS: Administration of ω-3FAs with gemcitabine chemotherapy in APC results in a significant decrease of MDSCs and stability of Tregs. This may be secondary to the reduction of pro-inflammatory mediators. A phase three randomized trial is justified to further examine these effects.

3.
J Knee Surg ; 33(9): 919-926, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31121632

ABSTRACT

As more commercial insurance companies adopt a bundled reimbursement model, similar to the Comprehensive Care for Joint Replacement (CJR) algorithm for Medicare beneficiaries, accurate risk adjustment of patient-reported outcomes (PROs) is critical to ensure success. With this movement toward bundled reimbursement, it is unknown if a formula adjusting for similar risks in the Medicare population could be applied to PROs in commercially insured and Medicare Advantage populations undergoing total knee arthroplasty (TKA). This study was performed to compare PROs after TKA in these insurance groups after adjusting for proposed risks. Demographics and clinical data were abstracted from medical records of 302 patients who underwent TKA performed by a single surgeon at a university-based orthopaedic practice during 2013 to 2017. Differences in PROs between commercially insured, Medicare Advantage, and Medicare patients during the 6 months following surgery were evaluated while controlling for demographics, clinical data, and baseline PRO scores. Medicare and Medicare Advantage patients were older (p < 0.001) and had more comorbidities (p = 0.001) than commercial patients. During the first 3 months following TKA, patients in all three groups experienced similar rates of recovery. At 6 months after surgery, outcomes began to diverge by insurance group. Medicare patients reported significantly less ability to perform activities of daily living (78.6 vs. 63.2; p = 0.001), worse physical function (39.6 vs. 44.9; p = 0.003), and more pain interference (57.9 vs. 52.4; p = 0.018) at day 180 than commercially insured patients. There were no statistically significant differences between Medicare Advantage patients and either commercially insured or Medicare patients. Therefore, commercial insurance companies that intend to apply a risk-adjusted equation similar to the CJR algorithm to commercial populations should be cautioned since the postoperative outcomes in this investigation differed after adjusting for the same risk factors that have been proposed for inclusion in the CJR algorithm. Nonetheless, further studies should be performed to ensure that companies participating in bundled reimbursement models have a positive influence on comprehensive health care for patients and providers. This is a level III, retrospective prognostic study.


Subject(s)
Arthroplasty, Replacement, Knee , Patient Reported Outcome Measures , Aged , Female , Humans , Insurance, Health , Male , Medicare , Medicare Part C , Middle Aged , Private Sector , Retrospective Studies , United States
4.
J Am Acad Orthop Surg Glob Res Rev ; 3(4): e062, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31334478

ABSTRACT

Tumoral calcinosis (TC) is an exceedingly rare disease, significantly so when located in the spine. Here, we present a 4-month-old patient with decreased head control and range of motion of the neck for several weeks. CT and MRI demonstrated a calcified mass in the retropharyngeal area and surrounding C1/C2, and TC was suspected. The patient underwent surgical biopsy and aspiration, which confirmed TC. The purpose of this case report is to document a rare disease, significantly so when taking into account both the location of the lesion and the patient's age, and to detail the treatment and response.

5.
J Nutr ; 149(7): 1133-1139, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31132111

ABSTRACT

BACKGROUND: Curcumin is the main active ingredient of the spice turmeric, investigated extensively for putative anticancer properties. OBJECTIVES: This phase IIa open-labelled randomized controlled trial aimed to assess safety, efficacy, quality of life, neurotoxicity, curcuminoids, and C-X-C-motif chemokine ligand 1 (CXCL1) in patients receiving folinic acid/5-fluorouracil/oxaliplatin chemotherapy (FOLFOX) compared with FOLFOX + 2 g oral curcumin/d (CUFOX). METHODS: Twenty-eight patients aged >18 y with a histological diagnosis of metastatic colorectal cancer were randomly assigned (1:2) to receive either FOLFOX or CUFOX. Safety was assessed by Common Toxicity Criteria-Adverse Event reporting, and efficacy via progression-free survival (PFS) and overall survival (OS). Quality of life and neurotoxicity were assessed using questionnaires (European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 and Functional Assessment of Cancer Treatment-Gynecologic Oncology Group-Neurotoxicity). Plasma curcuminoids were determined with liquid chromatography (LC) electrospray ionization tandem mass spectrometry and CXCL1 by ELISA. RESULTS: Addition of daily oral curcumin to FOLFOX chemotherapy was safe and tolerable (primary outcome). Similar adverse event profiles were observed for both arms. In the intention-to-treat population, the HR for PFS was 0.57 (95% CI: 0.24, 1.36; P = 0.2) (median of 171 and 291 d for FOLFOX and CUFOX, respectively) and for OS was 0.34 (95% CI: 0.14, 0.82; P = 0.02) (median of 200 and 502 d for FOLFOX and CUFOX, respectively). There was no significant difference between arms for quality of life (P = 0.248) or neurotoxicity (P = 0.223). Curcumin glucuronide was detectable at concentrations >1.00 pmol/mL in 15 of 18 patients receiving CUFOX. Curcumin did not significantly alter CXCL1 over time (P = 0.712). CONCLUSION: Curcumin is a safe and tolerable adjunct to FOLFOX chemotherapy in patients with metastatic colorectal cancer. This trial was registered at clinicaltrials.gov as NCT01490996 and at www.clinicaltrialsregister.eu as EudraCT 2011-002289-19.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Curcumin/therapeutic use , Administration, Oral , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colorectal Neoplasms/pathology , Curcumin/administration & dosage , Female , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Humans , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Metastasis , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Treatment Outcome
6.
Br J Cancer ; 119(1): 27-35, 2018 07.
Article in English | MEDLINE | ID: mdl-29925934

ABSTRACT

BACKGROUND: Advanced biliary tract cancer (ABC) has a poor prognosis. Cediranib, in addition to cisplatin/gemcitabine [CisGem], improved the response rate, but did not improve the progression-free survival (PFS) in the ABC-03 study. Minimally invasive biomarkers predictive of cediranib benefit may improve patient outcomes. METHODS: Changes in 15 circulating plasma angiogenesis or inflammatory-related proteins and cytokeratin-18 (CK18), measured at baseline and during therapy until disease progression, were correlated with overall survival (OS) using time-varying covariate Cox models (TVC). RESULTS: Samples were available from n = 117/124 (94%) patients. Circulating Ang1&2, FGFb, PDGFbb, VEGFC, VEGFR1 and CK18 decreased as a result of the therapy, independent of treatment with cediranib. Circulating VEGFR2 and Tie2 were preferentially reduced by cediranib. Patients with increasing levels of VEGFA at any time had a worse PFS and OS; this detrimental effect was attenuated in patients receiving cediranib. TVC analysis revealed CK18 and VEGFR2 increases correlated with poorer OS in all patients (P < 0.001 and P = 0.02, respectively). CONCLUSIONS: Rising circulating VEGFA levels in patients with ABC, treated with CisGem, are associated with worse PFS and OS, not seen in patients receiving cediranib. Rising levels of markers of tumour burden (CK18) and potential resistance (VEGFR2) are associated with worse outcomes and warrant validation.


Subject(s)
Biliary Tract Neoplasms/drug therapy , Keratin-18/blood , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-2/blood , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biliary Tract Neoplasms/blood , Biliary Tract Neoplasms/pathology , Biomarkers, Tumor/blood , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins/blood , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Quinazolines/administration & dosage , Treatment Outcome , United Kingdom , Gemcitabine
7.
Br J Cancer ; 118(6): 793-801, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29438372

ABSTRACT

BACKGROUND: The Notch pathway is frequently activated in cancer. Pathway inhibition by γ-secretase inhibitors has been shown to be effective in pre-clinical models of pancreatic cancer, in combination with gemcitabine. METHODS: A multi-centre, non-randomised Bayesian adaptive design study of MK-0752, administered per os weekly, in combination with gemcitabine administered intravenously on days 1, 8 and 15 (28 day cycle) at 800 or 1000 mg m-2, was performed to determine the safety of combination treatment and the recommended phase 2 dose (RP2D). Secondary and tertiary objectives included tumour response, plasma and tumour MK-0752 concentration, and inhibition of the Notch pathway in hair follicles and tumour. RESULTS: Overall, 44 eligible patients (performance status 0 or 1 with adequate organ function) received gemcitabine and MK-0752 as first or second line treatment for pancreatic cancer. RP2Ds of MK-0752 and gemcitabine as single agents could be combined safely. The Bayesian algorithm allowed further dose escalation, but pharmacokinetic analysis showed no increase in MK-0752 AUC (area under the curve) beyond 1800 mg once weekly. Tumour response evaluation was available in 19 patients; 13 achieved stable disease and 1 patient achieved a confirmed partial response. CONCLUSIONS: Gemcitabine and a γ-secretase inhibitor (MK-0752) can be combined at their full, single-agent RP2Ds.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Adult , Aged , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bayes Theorem , Benzene Derivatives/administration & dosage , Benzene Derivatives/adverse effects , Benzene Derivatives/pharmacokinetics , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Propionates/administration & dosage , Propionates/adverse effects , Propionates/pharmacokinetics , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Signal Transduction/drug effects , Sulfones/administration & dosage , Sulfones/adverse effects , Sulfones/pharmacokinetics , Gemcitabine
8.
JPEN J Parenter Enteral Nutr ; 41(3): 398-403, 2017 03.
Article in English | MEDLINE | ID: mdl-26220200

ABSTRACT

BACKGROUND: Marine-derived ω-3 fatty acids (ω-3FAs) have proven antitumor activity in vivo and in vitro and improve quality of life (QOL) in clinical cancer studies. These changes may be mediated by reduction in circulating proangiogenic and proinflammatory factors. In this first study of intravenous ω-3FAs as a therapy in cancer patients, we aimed to assess if it could augment the antitumor activity of gemcitabine in patients with advanced pancreatic cancer and improve QOL. MATERIALS AND METHODS: Patients were administered gemcitabine 1000 mg/m3 weekly followed by up to 100 g (200 mg/mL) of ω-3 rich lipid emulsion for 3 weeks followed by a rest week. This was continued for up to 6 cycles, progression, unacceptable toxicity, patient request, or death. The primary outcome measure was objective response rate, with secondary outcome measures of overall and progression free survival, QOL scores, and adverse events. RESULTS: Fifty patients were recruited. Response rate was 14.3% and disease control rate was 85.7%. Overall and progression free survival were 5.9 and 4.8 months, respectively. Increase in global health of > 10% over baseline was seen in 47.2% of patients. More than 50% of patients had > 10% increase in QOL scores in generic symptom scores and both disease-specific domains. Grade 3/4 adverse events were thrombocytopenia (8%), neutropenia (12%), nausea or vomiting (4%), and chills (6%). CONCLUSION: Intravenous ω-3FAs in combination with gemcitabine shows evidence of improved activity and benefit to QOL in patients with advanced pancreas cancer and is worthy of investigation in a randomized phase III trial.


Subject(s)
Administration, Intravenous , Deoxycytidine/analogs & derivatives , Fatty Acids, Omega-3/therapeutic use , Pancreatic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Deoxycytidine/therapeutic use , Disease-Free Survival , Drug Therapy, Combination , Endpoint Determination , Female , Humans , Male , Middle Aged , Quality of Life , Treatment Outcome , Gemcitabine
9.
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
10.
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
11.
Trials ; 16: 110, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25872567

ABSTRACT

BACKGROUND: The need for low toxicity adjuncts to standard care chemotherapy in inoperable colorectal cancer, with potential to improve outcomes and decrease the side-effect burden, is well recognised. Addition of the low toxicity diet-derived agent, curcumin (the active ingredient of turmeric), to standard oxaliplatin-based therapy has shown promise in numerous pre-clinical studies. METHODS/DESIGN: This study is the first to combine daily oral curcumin with standard care FOLFOX-based (5-fluorouracil, folinic acid and oxaliplatin) chemotherapy in colorectal cancer patients with inoperable liver metastases: the CUFOX trial. CUFOX comprises a Phase 1 dose-escalation study (3 + 3 + 3 design) to determine an acceptable target dose of curcumin with which to safely proceed to a Phase IIa open-labelled randomised controlled trial. Thirty three participants with histological or cytological confirmation of inoperable colorectal cancer will then be randomised to oxaliplatin-based chemotherapy with the addition of daily oral curcumin at the target dose determined in Phase I, or to standard care oxaliplatin-based chemotherapy alone (recruiting at a ratio of 2:1). DISCUSSION: Primary outcome measures will be the determination of a target dose which is both safe and tolerable for long-term administration to individuals in receipt of first-line oxaliplatin-based chemotherapy for inoperable colorectal cancer. Secondary outcome measures will include observation of any changes in neuropathic side-effects of chemotherapy, improvement to progression-free or overall survival and identification of putative efficacy biomarkers in plasma. The results will be disseminated via presentation at national and international conferences, via publication in appropriate peer-reviewed journals and via the Cancer Research UK/Department of Health Experimental Cancer Medicine Centre Network. This trial has full ethical and institutional approval, and commenced recruitment in February 2012. TRIAL REGISTRATION: ClinicalTrials.gov ( NCT01490996 , registered 7(th) December 2011), European Drug Regulating Authorities (EudraCT 2011-002289-19, registered 13(th) May 2011), UKCRN ID#10672.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Curcumin/administration & dosage , Liver Neoplasms/drug therapy , Administration, Oral , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/blood , Clinical Protocols , Colorectal Neoplasms/blood , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Curcumin/adverse effects , Disease Progression , Disease-Free Survival , Drug Administration Schedule , England , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Liver Neoplasms/blood , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Research Design , Time Factors , Treatment Outcome
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.
Eur J Cancer Prev ; 23(6): 594-601, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25259886

ABSTRACT

Information comparing attitudes towards taking cancer chemopreventive agents and assessing drug characteristics that would make chemopreventive agents more acceptable to participants is essential for future trial design and to ultimately promote compliance. We therefore undertook a cross-sectional questionnaire study, the aim of which was to assess current attitudes towards chemopreventive agents and to determine which characteristics make chemopreventive agents more acceptable to potential target populations. Questionnaires were distributed to four groups of participants: university students, cancer patients, partners/spouses of patients with cancer and individuals at a high familial risk for cancer. The survey's overall response rate was 35.5% (350 participants). The majority of participants (92.9%) considered taking cancer chemopreventive agents. Factors that positively influenced participants towards chemoprevention were a family history of cancer and having children. Diet-derived chemopreventive agents were preferred by 74.6%, who associated these agents with being 'healthier' and having a better side-effect profile. Most participants preferred either two medium-sized capsules or four small capsules daily. Overall, participants were keen to consider chemoprevention, particularly in cases in which cancer risk was high, and preferred diet-derived agents, believing them to have minimal side-effects.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Attitude to Health , Chemoprevention , Neoplasms/prevention & control , Adult , Aged , Aged, 80 and over , Chemoprevention/psychology , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Patient Acceptance of Health Care/psychology , Patient Acceptance of Health Care/statistics & numerical data , Risk , Surveys and Questionnaires , Young Adult
14.
Lancet Oncol ; 15(8): 829-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24954781

ABSTRACT

BACKGROUND: We aimed to assess the efficacy and safety of sequential or simultaneous telomerase vaccination (GV1001) in combination with chemotherapy in patients with locally advanced or metastatic pancreatic cancer. METHODS: TeloVac was a three-group, open-label, randomised phase 3 trial. We recruited patients from 51 UK hospitals. Eligible patients were treatment naive, aged older than 18 years, with locally advanced or metastatic pancreatic ductal adenocarcinoma, and Eastern Cooperative Oncology Group performance status of 0-2. Patients were randomly assigned (1:1:1) to receive either chemotherapy alone, chemotherapy with sequential GV1001 (sequential chemoimmunotherapy), or chemotherapy with concurrent GV1001 (concurrent chemoimmunotherapy). Treatments were allocated with equal probability by means of computer-generated random permuted blocks of sizes 3 and 6 in equal proportion. Chemotherapy included six cycles of gemcitabine (1000 mg/m(2), 30 min intravenous infusion, at days 1, 8, and 15) and capecitabine (830 mg/m(2) orally twice daily for 21 days, repeated every 28 days). Sequential chemoimmunotherapy included two cycles of combination chemotherapy, then an intradermal lower abdominal injection of granulocyte-macrophage colony-stimulating factor (GM-CSF; 75 µg) and GV1001 (0·56 mg; days 1, 3, and 5, once on weeks 2-4, and six monthly thereafter). Concurrent chemoimmunotherapy included giving GV1001 from the start of chemotherapy with GM-CSF as an adjuvant. The primary endpoint was overall survival; analysis was by intention to treat. This study is registered as an International Standard Randomised Controlled Trial, number ISRCTN4382138. FINDINGS: The first patient was randomly assigned to treatment on March 29, 2007, and the trial was terminated on March 27, 2011. Of 1572 patients screened, 1062 were randomly assigned to treatment (358 patients were allocated to the chemotherapy group, 350 to the sequential chemoimmunotherapy group, and 354 to the concurrent chemoimmunotherapy group). We recorded 772 deaths; the 290 patients still alive were followed up for a median of 6·0 months (IQR 2·4-12·2). Median overall survival was not significantly different in the chemotherapy group than in the sequential chemoimmunotherapy group (7·9 months [95% CI 7·1-8·8] vs 6·9 months [6·4-7·6]; hazard ratio [HR] 1·19, 98·25% CI 0·97-1·48, p=0·05), or in the concurrent chemoimmunotherapy group (8·4 months [95% CI 7·3-9·7], HR 1·05, 98·25% CI 0·85-1·29, p=0·64; overall log-rank of χ(2)2df=4·3; p=0·11). The commonest grade 3-4 toxic effects were neutropenia (68 [19%] patients in the chemotherapy group, 58 [17%] patients in the sequential chemoimmunotherapy group, and 79 [22%] patients in the concurrent chemoimmunotherapy group; fatigue (27 [8%] in the chemotherapy group, 35 [10%] in the sequential chemoimmunotherapy group, and 44 [12%] in the concurrent chemoimmunotherapy group); and pain (34 [9%] patients in the chemotherapy group, 39 [11%] in the sequential chemoimmunotherapy group, and 41 [12%] in the concurrent chemoimmunotherapy group). INTERPRETATION: Adding GV1001 vaccination to chemotherapy did not improve overall survival. New strategies to enhance the immune response effect of telomerase vaccination during chemotherapy are required for clinical efficacy. FUNDING: Cancer Research UK and KAEL-GemVax.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/administration & dosage , Pancreatic Ducts , Pancreatic Neoplasms/drug therapy , Peptide Fragments/administration & dosage , Telomerase/administration & dosage , Adenocarcinoma/secondary , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cancer Vaccines/adverse effects , Capecitabine , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Fatigue/chemically induced , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Immunologic Factors/administration & dosage , Intention to Treat Analysis , Kaplan-Meier Estimate , Male , Middle Aged , Neutropenia/chemically induced , Pain/chemically induced , Pancreatic Neoplasms/pathology , Peptide Fragments/adverse effects , T-Lymphocytes/immunology , Telomerase/adverse effects , Gemcitabine
15.
J Pharmacol Exp Ther ; 350(3): 483-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24939419

ABSTRACT

Lung cancer is responsible for over one million deaths worldwide each year. Smoking cessation for lung cancer prevention remains key, but it is increasingly acknowledged that prevention strategies also need to focus on high-risk groups, including ex-smokers, and patients who have undergone resection of a primary tumor. Models for chemoprevention of lung cancer often present conflicting results, making rational design of lung cancer chemoprevention trials challenging. There has been much focus on use of dietary bioactive compounds in lung cancer prevention strategies, primarily due to their favorable toxicity profile and long history of use within the human populace. One such compound is curcumin, derived from the spice turmeric. This review summarizes and stratifies preclinical evidence for chemopreventive efficacy of curcumin in models of lung cancer, and adjudges the weight of evidence for use of curcumin in lung cancer chemoprevention strategies.


Subject(s)
Curcumin/administration & dosage , Curcumin/therapeutic use , Evidence-Based Medicine/methods , Lung Neoplasms/prevention & control , Primary Prevention/methods , Secondary Prevention/methods , Tertiary Prevention/methods , Translational Research, Biomedical/methods , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Cell Line, Tumor , Curcumin/chemistry , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/epidemiology , Smoking Cessation/methods
16.
Int J Cancer ; 134(5): 1102-11, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-23959905

ABSTRACT

Bladder cancer patients suffer significant treatment failure, including high rates of recurrence and poor outcomes for advanced disease. If mechanisms to improve tumour cell treatment sensitivity could be identified and/or if tumour response could be predicted, it should be possible to improve local-control and survival. Previously, we have shown that radiation-induced DNA damage, measured by alkaline Comet assay (ACA), correlates bladder cancer cell radiosensitivity in vitro. In this study we first show that modified-ACA measures of cisplatin and mitomycin-C-induced damage also correlate bladder cancer cell chemosensitivity in vitro, with essentially the same rank order for chemosensitivity as for radiosensitivity. Furthermore, ACA studies of radiation-induced damage in different cell-DNA substrates (nuclei, nucleoids and intact parent cells) suggest that it is a feature retained in the prepared nucleoids that is responsible for the relative damage sensitivity of bladder cancer cells, suggestive of differences in the organisation of DNA within resistant vs. sensitive cells. Second, we show that ACA analysis of biopsies from bladder tumours reveal that reduced DNA damage sensitivity associates with poorer treatment outcomes, notably that tumours with a reduced damage response show a significant association with local recurrence of non-invasive disease and that reduced damage response was a better predictor of recurrence than the presence of high-risk histology in this cohort. In conclusion, this study demonstrates that mechanisms governing treatment-induced DNA damage are both central to and predictive of bladder cancer cell treatment sensitivity and exemplifies a link between DNA damage resistance and both treatment response and tumour aggression.


Subject(s)
Comet Assay/methods , DNA Damage , Urinary Bladder Neoplasms/drug therapy , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Mitomycin/pharmacology , Treatment Outcome , Urinary Bladder Neoplasms/genetics
17.
JPEN J Parenter Enteral Nutr ; 38(2): 214-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23423329

ABSTRACT

BACKGROUND: Pancreatic cancer has an extremely poor clinical outcome. Surrogate biomarkers for outcome are scarce. There is mixed evidence for the association of high mannose-binding lectin (MBL) complement activity with cancer outcomes, including reduced survival and increased infectious complications. ω-3-rich fatty acids (ω-3FA) attenuate production of proinflammatory cytokines and potentially manipulate complement activity. MATERIALS AND METHODS: As part of a single-arm phase II trial in a university hospital, patients with advanced pancreatic adenocarcinoma were treated with weekly ω-3FA-rich intravenous infusion (Lipidem [B. Braun Melsungen AG, Melsungen, Germany]: up to 100 g/wk) plus gemcitabine chemotherapy until withdrawal or tumor progression. Primary outcome measure was objective response rate. Changes in complement activity, which were a secondary outcome measure, were analyzed and relation to clinical outcome determined. RESULTS: Twenty-three patients were assessable for time to progression (TTP), overall survival (OS), and complement activity. No hypoactivity in alternative and classical pathways was demonstrated. Baseline MBL was low in 10 of 23 patients (43.5%). There was no difference in OS or TTP between low- and high-baseline MBL patients. Of these 10 patients, 5 were classified as MBL responders. MBL responders had a tendency toward improved OS over nonresponders (8.9 vs 4.4 months, P = .07). MBL responders had significantly improved ttp over nonresponders (10.6 VS 5.3 MONTHS, P = .03). CONCLUSION: MBL restoration had an association with improved outcome in the cohort of patients with low MBL activity at baseline. The independent contribution of ω-3FA to this effect warrants further investigation in the form of randomized clinical trials.


Subject(s)
Complement System Proteins/metabolism , Deoxycytidine/analogs & derivatives , Fatty Acids, Omega-3/therapeutic use , Fish Oils/therapeutic use , Mannose-Binding Lectin/blood , Pancreatic Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Cytokines/blood , Deoxycytidine/therapeutic use , Female , Humans , Male , Mannose-Binding Lectin/metabolism , Middle Aged , Gemcitabine
18.
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
19.
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
20.
Ann N Y Acad Sci ; 1290: 12-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23855461

ABSTRACT

Among the plethora of biochemical mechanisms engaged by resveratrol in preclinical systems, its anticarcinogenic effects represent some of the most convincing and intriguing. As outlined in this review, there is considerable interest in developing resveratrol for cancer prevention and treatment. The plasma pharmacokinetics of resveratrol in humans are now reasonably well defined, and studies have shown that repeated daily doses up to 1 g are safe and well tolerated, although gastrointestinal toxicity is observed at higher intakes. However, care is needed regarding underlying conditions in specific patient groups, and there is potential for drug interactions at doses greater than 1 gram. Little is known regarding the pharmacodynamic effects of resveratrol in humans, but the observation that it modulates components of the insulin-like growth factor system in the plasma of volunteers is encouraging. While the knowledge base that helps determine whether resveratrol may be useful in cancer management has increased substantially in recent years, important questions remain.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Stilbenes/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Clinical Trials as Topic/methods , Disease Management , Dose-Response Relationship, Drug , Humans , Neoplasms/metabolism , Resveratrol , Stilbenes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...