Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Br J Cancer ; 128(2): 297-309, 2023 01.
Article in English | MEDLINE | ID: mdl-36347967

ABSTRACT

INTRODUCTION: Colorectal cancer is the fourth most common cancer in the UK. There remains a need for improved risk stratification following curative resection. Circulating-tumour DNA (ctDNA) has gained particular interest as a cancer biomarker in recent years. We performed a systematic review to assess the utility of ctDNA in identifying minimal residual disease in colorectal cancer. METHODS: Studies were included if ctDNA was measured following curative surgery and long-term outcomes were assessed. Studies were excluded if the manuscript could not be obtained from the British Library or were not available in English. RESULTS: Thirty-seven studies met the inclusion criteria, involving 3002 patients. Hazard ratios (HRs) for progression-free survival (PFS) were available in 21 studies. A meta-analysis using a random effects model demonstrated poorer PFS associated with ctDNA detection at the first liquid biopsy post-surgery [HR: 6.92 CI: 4.49-10.64 p < 0.00001]. This effect was also seen in subgroup analysis by disease extent, adjuvant chemotherapy and assay type. DISCUSSION: Here we demonstrate that ctDNA detection post-surgery is associated with a greater propensity to disease relapse and is an independent indicator of poor prognosis. Prior to incorporation into clinical practice, consensus around timing of measurements and assay methodology are critical. PROTOCOL REGISTRATION: The protocol for this review is registered on PROSPERO (CRD42021261569).


Subject(s)
Colorectal Neoplasms , Neoplasm Recurrence, Local , Humans , Neoplasm, Residual/genetics , Neoplasm Recurrence, Local/pathology , Chemotherapy, Adjuvant , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/surgery , Biomarkers, Tumor/genetics
2.
BMC Cancer ; 23(1): 820, 2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37667231

ABSTRACT

BACKGROUND: Global annual cancer incidence is forecast to rise to 27.5 M by 2040, a 62% increase from 2018. For most cancers, prevention and early detection are the most effective ways of reducing mortality. This study maps trials in cancer screening, prevention, and early diagnosis (SPED) to identify areas of unmet need and highlight research priorities. METHODS: A systematic mapping review was conducted to evaluate all clinical trials focused on cancer SPED, irrespective of tumour type. The National Cancer Research Institute (NCRI) portfolio, EMBASE, PubMed and Medline were searched for relevant papers published between 01/01/2007 and 01/04/2020. References were exported into Covidence software and double-screened. Data were extracted and mapped according to tumour site, geographical location, and intervention type. RESULTS: One hundred seventeen thousand seven hundred one abstracts were screened, 5157 full texts reviewed, and 2888 studies included. 1184 (52%) trials focussed on screening, 554 (24%) prevention, 442 (20%) early diagnosis, and 85 (4%) a combination. Colorectal, breast, and cervical cancer comprised 61% of all studies compared with 6.4% in lung and 1.8% in liver cancer. The latter two are responsible for 26.3% of global cancer deaths compared with 19.3% for the former three. Number of studies varied markedly according to geographical location; 88% were based in North America, Europe, or Asia. CONCLUSIONS: This study shows clear disparities in the volume of research conducted across different tumour types and according to geographical location. These findings will help drive future research effort so that resources can be directed towards major challenges in cancer SPED.


Subject(s)
Liver Neoplasms , Uterine Cervical Neoplasms , Female , Humans , Early Detection of Cancer , Asia , Breast
3.
Br J Nurs ; 31(10): S14-S24, 2022 May 26.
Article in English | MEDLINE | ID: mdl-35648663

ABSTRACT

More people in the UK are living with cancer than ever before. With an increasingly ethnically diverse population, greater emphasis must be placed on understanding factors influencing cancer outcomes. This review seeks to explore UK-specific variations in engagement with cancer services in minority ethnic groups and describe successful interventions. The authors wish to highlight that, despite improvement to engagement and education strategies, inequalities still persist and work to improve cancer outcomes across our communities still needs to be prioritised. There are many reasons why cancer healthcare inequities exist for minority communities, reported on a spectrum ranging from cultural beliefs and awareness, through to racism. Strategies that successfully enhanced engagement included language support; culturally-sensitive reminders; community-based health workers and targeted outreach. Focusing on the diverse city of Leicester the authors describe how healthcare providers, researchers and community champions have worked collectively, delivering targeted community-based strategies to improve awareness and access to cancer services.


Subject(s)
Minority Groups , Neoplasms , Early Detection of Cancer , Ethnic and Racial Minorities , Ethnicity , Humans , Neoplasms/diagnosis , United Kingdom
4.
Nutr Metab Cardiovasc Dis ; 31(1): 14-22, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33223399

ABSTRACT

AIM: Whether the relative risk of cancer incidence and mortality associated with diabetes has changed over time is unknown. DATA SYNTHESIS: On August 12th, 2020, we electronically searched for observational studies reporting on the association between diabetes and cancer. We estimated temporal trends in the relative risk of cancer incidence or mortality associated with diabetes and calculated the ratio of relative risk (RRR) comparing different periods. As many as 193 eligible articles, reporting data on 203 cohorts (56,852,381 participants; 3,735,564 incident cancer cases; 185,404 cancer deaths) and covering the period 1951-2013, were included. The relative risk of all-site cancer incidence increased between 1980 and 2000 [RRR 1990 vs.1980: (1.24; 95% CI: 1.16, 1.34); 2000 vs.1990: (1.23; 1.15, 1.31)] and stabilised thereafter at a relative risk of 1.2; the relative risk of all-site cancer mortality was constant at about 1.2 from 1980 to 2010. Both magnitudes and trends in relative risk varied across cancer sites: the relative risk of colorectal, female breast, and endometrial cancer incidence and pancreatic cancer mortality was constant during the observed years; it increased for bladder, stomach, kidney, and pancreatic cancer incidence until 2000; and decreased for liver while increased for prostate, colon and gallbladder cancer incidence after 2000. CONCLUSIONS: Alongside the increasing prevalence of diabetes, the temporal patterns of the relative risk of cancer associated with diabetes may have contributed to the current burden of cancer in people with diabetes.


Subject(s)
Diabetes Mellitus/mortality , Neoplasms/mortality , Cause of Death/trends , Diabetes Mellitus/diagnosis , Humans , Incidence , Neoplasms/diagnosis , Observational Studies as Topic , Prevalence , Risk Assessment , Risk Factors , Time Factors
5.
Molecules ; 25(2)2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31963196

ABSTRACT

Curcumin has been investigated extensively for cancer prevention, but it has been proposed that long-term treatments may promote clonal evolution and gain of cellular resistance, potentially rendering cancer cells less sensitive to future therapeutic interventions. Here, we used long-term, low-dose treatments to determine the potential for adverse effects in non-small cell lung cancer (NSCLC) cells. IC50s for curcumin, cisplatin, and pemetrexed in A549, PC9, and PC9ER NSCLC cells were evaluated using growth curves. IC50s were subsequently re-assessed following long-term, low-dose curcumin treatment and a three-month treatment withdrawal period, with a concurrent assessment of oncology-related protein expression. Doublet cisplatin/pemetrexed-resistant cell lines were created and the IC50 for curcumin was determined. Organotypic NSCLC-fibroblast co-culture models were used to assess the effects of curcumin on invasive capacity. Following long-term treatment/treatment withdrawal, there was no significant change in IC50s for the chemotherapy drugs, with chemotherapy-resistant cell lines exhibiting similar sensitivity to curcumin as their non-resistant counterparts. Curcumin (0.25-0.5 µM) was able to inhibit the invasion of both native and chemo-resistant NSCLC cells in the organotypic co-culture model. In summary, long-term curcumin treatment in models of NSCLC neither resulted in the acquisition of pro-carcinogenic phenotypes nor caused resistance to chemotherapy agents.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Fibroblasts , Humans , Immunohistochemistry , Mice , Time Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
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
7.
Lipids Health Dis ; 17(1): 19, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29378575

ABSTRACT

BACKGROUND: There is limited evidence assessing the effects of omega-3 polyunsaturated fatty acids (PUFAs) on oesophageal adenocarcinoma, both in vitro and in vivo. We evaluated the effects of the omega-3 PUFA and oxaliplatin on OE33 and OE19 cells. METHOD: The two oesophageal cells were treated with Omegaven® (fish oil emulsion), EPA, DHA and oxaliplatin and incubated for up to 144 h. RESULTS: The following inhibitory effects were observed on OE33 cells: EPA reduced cell growth by 39% (p = 0.001), DHA by 59% (p < 0.000) and Oxaliplatin by 77% (p < 0.000). For OE19 cells, the EPA reduced growth by 1% (p = 0.992), DHA by 26% (p = 0.019) and oxaliplatin by 76% (p < 0.000). For both cells, Omegaven® resulted in reduced cell growth at intermediate concentrations (20-40 µM) and increased cell growth at low (10 µM) and high (50 µM) concentrations. DHA, Omegaven® and oxaliplatin were associated with significant downregulation of VEGF and p53 protein, and upregulation of p21 protein. DHA, Omegaven® and Oxaliplatin also led to significant downregulation of the total ERK1/2 and Akt proteins. CONCLUSION: DHA, Omegaven® and oxaliplatin were associated with downregulation of p53 and VEGF in both cells. Of the PUFAs studied, DHA alone or in combination (Omegaven®) had greater in vitro anti-cancer effects than EPA alone.


Subject(s)
Adenocarcinoma/drug therapy , Esophageal Neoplasms/drug therapy , Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Organoplatinum Compounds/pharmacology , Signal Transduction/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/physiopathology , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/drug effects , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Down-Regulation , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/physiopathology , Fatty Acids, Omega-3/therapeutic use , Female , Fish Oils/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Male , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Triglycerides , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factors/drug effects , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
8.
Biomed Chromatogr ; : e4280, 2018 May 09.
Article in English | MEDLINE | ID: mdl-29744910

ABSTRACT

Curcumin, derived from turmeric, has been extensively investigated for its broad spectrum of biological activities. Previously reported HPLC-UV methods have focussed on analysis of the parent compound. Here, a sensitive HPLC-UV method was developed and partially validated, then used for the simultaneous determination of curcumin and its glucuronide and sulfate metabolites in plasma and lung tissue from mice. The assay was applied to an in vivo pharmacokinetic study comparing formulated curcumin (Meriva™) with standard curcumin. Plasma levels of glucuronide and sulfate metabolites were 5- and 2-fold higher after Meriva™ administration compared with standard curcumin. In lung tissue, free curcumin was 4-fold higher following Meriva™ administration vs standard curcumin. This assay represents a rapid, cheap method for simultaneous detection of curcumin and its major metabolites that has applicability in pre-clinical settings.

9.
Int J Cancer ; 138(1): 30-44, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-25611701

ABSTRACT

The tumour microenvironment plays an essential role in the development and spread of cancers. Tumour cells interact with the surrounding extracellular matrix (ECM), embedded within which, are a variety of non-cancer cells including cells of the vasculature, immune system and fibroblasts. The essential role of fibroblasts in the cultivation and maintenance of an environment in which tumour cells are able to maintain their aggressive phenotypic traits is becoming increasingly well documented. Cancer-associated fibroblasts are able to secrete a vast array of ECM-modulating factors, meaning that they have potential for a functional role in every step of the carcinogenic process. In particular, they are likely to have a role in early tumour-initiating inflammatory events, and so may provide a potential target for chemopreventive intervention. This review summarises the known interactions between lung tumour cells and surrounding reactive fibroblasts, highlighting the need to further investigate cancer-associated fibroblasts as therapeutic targets in lung cancer chemoprevention strategies.


Subject(s)
Carcinogenesis , Fibroblasts/metabolism , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Stromal Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Communication , Chemoprevention , Drug Resistance, Neoplasm , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Molecular Targeted Therapy , Prognosis , Signal Transduction/drug effects , Stem Cells/metabolism , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
10.
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
11.
PLoS One ; 17(2): e0262364, 2022.
Article in English | MEDLINE | ID: mdl-35130302

ABSTRACT

Research into the metabolism of the non-essential amino acid (NEAA) proline in cancer has gained traction in recent years. The last step in the proline biosynthesis pathway is catalyzed by pyrroline-5-carboxylate reductase (PYCR) enzymes. There are three PYCR enzymes: mitochondrial PYCR1 and 2 and cytosolic PYCR3 encoded by separate genes. The expression of the PYCR1 gene is increased in numerous malignancies and correlates with poor prognosis. PYCR1 expression sustains cancer cells' proliferation and survival and several mechanisms have been implicated to explain its oncogenic role. It has been suggested that the biosynthesis of proline is key to sustain protein synthesis, support mitochondrial function and nucleotide biosynthesis. However, the links between proline metabolism and cancer remain ill-defined and are likely to be tissue specific. Here we use a combination of human dataset, human tissue and mouse models to show that the expression levels of the proline biosynthesis enzymes are significantly increased during colorectal tumorigenesis. Functionally, the expression of mitochondrial PYCRs is necessary for cancer cells' survival and proliferation. However, the phenotypic consequences of PYCRs depletion could not be rescued by external supplementation with either proline or nucleotides. Overall, our data suggest that, despite the mechanisms underlying the role of proline metabolism in colorectal tumorigenesis remain elusive, targeting the proline biosynthesis pathway is a suitable approach for the development of novel anti-cancer therapies.


Subject(s)
Colorectal Neoplasms
12.
Int J Cancer ; 129(2): 476-86, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-20839263

ABSTRACT

The aims of this study were to determine potency of oxaliplatin in combination with curcumin in oxaliplatin-resistant cell lines in vitro and to evaluate the efficacy of a novel curcumin formulation (Meriva®) alone and in combination with oxaliplatin in colorectal tumor-bearing mice, exploring relevant pharmacodynamic markers in vivo. Oxaliplatin-resistant HCT116 p53wt and p53(-/-) cell lines were generated, and the effects of oxaliplatin in combination with curcumin on resistance- and proliferation-associated proteins investigated. Eighty nude mice were implanted with HCT116 p53wt colorectal cancer cells before randomization into the following treatment groups: control; Meriva only; oxaliplatin only; Meriva + oxaliplatin. Tumor volume was assessed, as was the expression of Ki-67, cleaved caspase-3 and Notch-1. Curcumin in combination with oxaliplatin was able to decrease proliferative capacity of oxaliplatin-resistant p53 wildtype and p53(-/-) cell lines more effectively than oxaliplatin alone. It also decreased markers associated with proliferation. After 21 days of treatment in the xenograft model, the order of efficacy was combination > Meriva > oxaliplatin > control. The decrease in tumor volume when compared to vehicle-treated animals was 53, 35 and 16%, respectively. Ki-67 and Notch-1 immunoreactivity was decreased by the combination when compared to vehicle-treated animals, with cleaved caspase-3 rising by 4.4-fold. Meriva did not adversely affect the DNA-platinating ability of oxaliplatin. Curcumin enhanced the cytotoxicity of oxaliplatin in models of oxaliplatin resistance in vitro. In vivo, Meriva greatly enhanced oxaliplatin efficacy, without affecting the mode of action of oxaliplatin. Addition of formulated curcumin to oxaliplatin-based chemotherapy regimens has the potential for clinical benefit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , HCT116 Cells/drug effects , Animals , Curcumin/pharmacology , Drug Synergism , Female , Humans , Mice , Mice, Nude , Organoplatinum Compounds/pharmacology , Oxaliplatin , Random Allocation , Xenograft Model Antitumor Assays
13.
Mol Nutr Food Res ; 62(16): e1800267, 2018 08.
Article in English | MEDLINE | ID: mdl-29943914

ABSTRACT

SCOPE: Curcumin (from turmeric), has been extensively investigated for potential beneficial properties in numerous diseases. Most work has focused on supra-dietary concentrations/doses that would necessitate curcumin supplementation. However, much evidence instigating curcumin research is underpinned by epidemiological data based on low dietary intake via turmeric consumption. METHODS AND RESULTS: Here, a novel, highly sensitive liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS/MS) method for detection of curcuminoids is described. Assay sensitivity is demonstrated in a pilot pharmacokinetic volunteer study following ingestion of foodstuffs containing a standardized mass of turmeric, representative of daily consumption by certain South Asian populations. Free parent curcumin was detectable in plasma from one individual, reaching maximal plasma concentrations (Cmax ) of 3.2 nm. Curcumin conjugates were detected in all volunteers; Cmax for curcumin glucuronide is 47.6 ± 28.5 nm 30 min post-food, while Cmax for demethoxycurcumin glucuronide and curcumin sulfate is ≈2 nm. Curcumin and its major metabolites persist in plasma for at least 8 h. CONCLUSION: Despite poor absorption and rapid conjugation, dietary intake of standard culinary turmeric within complex food matrices furnished human plasma with detectable levels of curcuminoids. Whether sustained low systemic concentrations of these non-nutritive, biologically active, dietary components may have pharmacological activity for human health benefit, warrants further research.


Subject(s)
Curcuma , Curcumin/analogs & derivatives , Curcumin/analysis , Glucuronides/blood , Adult , Calibration , Chromatography, Liquid , Curcumin/metabolism , Diarylheptanoids , Female , Food Analysis , Healthy Volunteers , Humans , Limit of Detection , Male , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
14.
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
15.
Cell Death Dis ; 9(9): 894, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30166531

ABSTRACT

Improving early detection of colorectal cancer (CRC) is a key public health priority as adenomas and stage I cancer can be treated with minimally invasive procedures. Population screening strategies based on detection of occult blood in the feces have contributed to enhance detection rates of localized disease, but new approaches based on genetic analyses able to increase specificity and sensitivity could provide additional advantages compared to current screening methodologies. Recently, circulating cell-free DNA (cfDNA) has received much attention as a cancer biomarker for its ability to monitor the progression of advanced disease, predict tumor recurrence and reflect the complex genetic heterogeneity of cancers. Here, we tested whether analysis of cfDNA is a viable tool to enhance detection of colon adenomas. To address this, we assessed a cohort of patients with adenomas and healthy controls using droplet digital PCR (ddPCR) and mutation-specific assays targeted to trunk mutations. Additionally, we performed multiregional, targeted next-generation sequencing (NGS) of adenomas and unmasked extensive heterogeneity, affecting known drivers such as APC, KRAS and mismatch repair (MMR) genes. However, tumor-related mutations were undetectable in patients' plasma. Finally, we employed a preclinical mouse model of Apc-driven intestinal adenomas and confirmed the inability to identify tumor-related alterations via cfDNA, despite the enhanced disease burden displayed by this experimental cancer model. Therefore, we conclude that benign colon lesions display extensive genetic heterogeneity, that they are not prone to release DNA into the circulation and are unlikely to be reliably detected with liquid biopsies, at least with the current technologies.


Subject(s)
Adenoma/diagnosis , Circulating Tumor DNA/isolation & purification , Colonic Neoplasms/diagnosis , Early Detection of Cancer/methods , Adenoma/blood , Adenoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Colonic Neoplasms/blood , Colonic Neoplasms/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Liquid Biopsy/methods , Male , Mice , Mice, Knockout , Middle Aged , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics
16.
Eur J Cancer Prev ; 26(4): 277-284, 2017 07.
Article in English | MEDLINE | ID: mdl-27128580

ABSTRACT

Familial adenomatous polyposis coli (FAP) is an autosomal dominant condition caused by a germline mutation in the adenomatous polyposis coli gene. Colonic adenomas form and almost all patients will develop colorectal cancer if they are not managed at an early stage. The safest preventive strategy is surgical resection of the colon, most commonly performed in late teenage years. There is a paucity of trials investigating the use of primary chemoprevention to delay polyp formation in paediatric FAP. There are extensive preclinical and early clinical data demonstrating that curcumin may be a safe and effective chemotherapeutic agent in reducing the polyp burden in this disease. We ultimately proposed to design and conduct a clinical study to assess whether curcumin treatment delays the need for surgery and/or prevents cancer in young patients with FAP. Research into clinical trial protocols has demonstrated that assessing patients' perceptions at the initial stage leads to better outcomes. We therefore conducted a questionnaire study of patients and parents of children affected by FAP to gain information to aid the protocol design. Results demonstrated that there are some FAP patients for whom this study is relevant and desirable. Those with a personal history of curcumin use reported that it was well tolerated. However, the response rate was poor (25%), indicating that there are potential difficulties ensuring adequate recruitment to the proposed trial. This report draws on lessons learnt from prior trials and the findings from the questionnaire to outline the challenges faced in designing such a study.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , Research Design/standards , Adolescent , Adult , Aged , Chemoprevention , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Surveys and Questionnaires , Young Adult
17.
Clin Cancer Res ; 11(23): 8521-7, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322316

ABSTRACT

PURPOSE AND EXPERIMENTAL DESIGN: Indole-3-carbinol has been proposed to induce apoptosis via a mechanism involving inhibition of protein kinase B (PKB) signaling in breast and prostate tumor cell lines. However, no functional data exist, and the effect of indole-3-carbinol on viability is known to be highly cell type specific. Here, we examine any requirement for PKB inhibition in induction of apoptosis by indole-3-carbinol in the MDA MB468 cell line using in vitro kinase assays, transfection, Western blotting, and flow cytometry. Comparison is also made with MCF10CA1 breast and PC3 prostate tumor cells. RESULTS: Indole-3-carbinol directly inhibited activity of phosphatidylinositol 3-kinase (PI3K) immunoprecipitated from HBL100 or MDA MB468 cells in vitro. Nonetheless, we present three lines of evidence that inhibition of PI3K/PKB signaling is not required for induction of apoptosis by indole-3-carbinol. First, 50% inhibition of PKB phosphorylation by LY294002 resulted in only 15% apoptosis after 72 hours, whereas similar PKB inhibition by indole-3-carbinol coincided with 30% apoptosis after only 24 hours. Second, induction of phospho-PKB (p-PKB) levels following stimulation with epidermal growth factor did not prevent indole-3-carbinol-induced apoptosis. Third, overexpression of active PKBalpha did not prevent induction of apoptosis by indole-3-carbinol. Inhibition of PKB phosphorylation by LY294002 in the PC3 and MCF10CA1 tumor cell lines similarly failed to result in a significant increase in apoptosis. CONCLUSIONS: Our results show that inhibition of PI3K/PKB signaling by indole-3-carbinol or LY294002 is not directly correlated with induction of apoptosis in several breast or prostate cell lines.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Indoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Chromones/pharmacology , Drug Combinations , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Female , Flow Cytometry , Humans , Immunoprecipitation , Male , Morpholines/pharmacology , Phosphorylation/drug effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Transfection , Tumor Cells, Cultured
18.
Mol Nutr Food Res ; 60(6): 1437-47, 2016 06.
Article in English | MEDLINE | ID: mdl-26603273

ABSTRACT

SCOPE: Pancreatic cancer remains a disease of poor prognosis, with alternate strategies being sought to improve therapeutic efficacy. Omega-3 fatty acids have shown clinical benefit, and mechanisms of action are under investigation. METHODS AND RESULTS: Proliferation assays, flow cytometry, invasion assays, ELISA and western blotting were used to investigate efficacy of omega-3 fatty acids alone and in combination with gemcitabine. The docosahexanoic acid (DHA)/eicosapentanoic acid (EPA) combination, Lipidem™, in combination with gemcitabine inhibited growth in pancreatic cancer and pancreatic stellate cell (PSC) lines, with PSCs exhibiting greatest sensitivity to this combination. Invasion of pancreatic cancer cells and PSCs in a 3D spheroid model, was inhibited by combination of gemcitabine with Lipidem™. PSCs were required for cancer cell invasion in an organotypic co-culture model, with invasive capacity reduced by Lipidem™ alone. Platelet-derived growth factor (PDGF) is a key cytokine in pro-proliferative and invasion signalling, and thus a critical regulator of interactions between pancreatic cancer cells and adjacent stroma. Platelet-derived growth factor (PDGF-BB) secretion was completely inhibited by the combination of Lipidem™ with gemcitabine in cancer cells and PSCs. CONCLUSION: Lipidem™ in combination with gemcitabine, has anti-proliferative and anti-invasive efficacy in vitro, with pancreatic stellate cells exhibiting the greatest sensitivity to this combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Fatty Acids, Omega-3/pharmacology , Pancreatic Neoplasms/drug therapy , Becaplermin , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Deoxycytidine/pharmacology , Humans , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Receptors, Platelet-Derived Growth Factor/metabolism , Gemcitabine
19.
Mol Cancer Ther ; 1(13): 1161-72, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12479697

ABSTRACT

We have identified a new target for the chemopreventive dietary agent indole-3-carbinol (13C) in the antiapoptotic signaling pathway involving phosphatidylinositol 3'-kinase and protein kinase B (PKB)/Akt. 13C inhibited phosphorylation and activation of PKB in the tumor-derived breast cell line MDA MB468, but not in the immortalized breast line HBL100. We propose that this cell type-specific response to 13C contributes to the differential induction of apoptosis and sensitivity to growth inhibition of the two cell lines (approximate IC50 = 30 microM for the MDA MB468 line, compared with 120 microM for the HBL100 line). 13C only induced apoptosis in the MDA MB468 cell line, but at higher doses, it increased necrosis in the HBL100 line. The tumor cell line was also markedly less able to recover when 13C was removed from the culture medium. Downstream of PKB, 13C decreased nuclear factor kappaB DNA binding, independently of an effect on IkappaB kinase, in the MDA MB468 cell line only. The tumor suppressor PTEN, which prevents phosphorylation and activation of PKB, was expressed in HBL100 cells but was not detected in MDA MB468 cells. In corroboration of the results obtained with the breast cell lines, 13C decreased phospho-PKB levels and induced apoptosis in the prostate cell line LNCaP, which expresses very low levels of PTEN, but did not do so in PTEN-positive DU145 cells. 13C did not affect PTEN levels in any cell line. This is the first study to report a differential mechanistic response of tumor-derived and nontumorigenic cell lines and of PTEN high- and low-expressing cells to 13C and indicates a promising chemopreventive role for 13C against estrogen receptor-alpha-negative, aggressive-phenotype breast tumors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Indoles/pharmacology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Breast/cytology , Breast Neoplasms/enzymology , Cell Division/drug effects , DNA, Neoplasm/metabolism , Female , Humans , I-kappa B Kinase , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylserines/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein , bcl-X Protein
20.
Cancer Chemother Pharmacol ; 76(1): 179-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26017621

ABSTRACT

PURPOSE: TMFol (3',4',5'-trimethoxyflavonol) is a synthetic analogue of the naturally occurring flavonol fisetin and quercetin, which have been considered of potential usefulness in the management of prostate cancer. We investigated whether TMFol may have preclinical features superior to those of its two flavonol congeners. METHODS: The ability of the three flavonols to compromise prostate cancer cell survival was tested in four prostate cancer cell types 22Rv1, TRAMP C2, PC-3 and LNCaP. The effect of TMFol on prostate cancer development in vivo was investigated in nude mice bearing the 22Rv1 or TRAMP C2 tumours. RESULTS: TMFol inhibited cell growth in vitro in all four prostate cancer cell types more potently than fisetin and quercetin. It also interfered with TRAMP C2 tumour development in vivo, while fisetin and quercetin at equivalent doses were without activity in this model. Likewise, TMFol slowed the growth of the 22Rv1 tumour in vivo. Efficacy in either model was accompanied by induction of apoptosis, although in vitro only TRAMP C2 cells, but not 22Rv1, underwent apoptosis when exposed to TMFol. CONCLUSIONS: The results support the notion that among the three congeneric flavonols, quercetin, fisetin and TMFol, the latter may be the most suitable candidate agent for potential development in prostate cancer management.


Subject(s)
Flavonols/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Humans , Male , Mice , Mice, Nude , Middle Aged , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL