+ All Categories
Home > Documents > Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 ›...

Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 ›...

Date post: 28-Jun-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
15
Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer A J Cockbain, 1 G J Toogood, 2 M A Hull 1 ABSTRACT Omega (u)-3 polyunsaturated fatty acids (PUFAs) are naturally occurring substances that are well tolerated and have been used extensively for the prevention of cardiovascular disease. More recently, u-3 PUFAs have been recognised to have anticancer activity. There is also evidence suggesting improved efficacy and/or tolerability of conventional cancer chemotherapy when administered with u-3 PUFAs. The purpose of this review is to (i) describe the mechanisms by which u-3 PUFAs are thought to have antineoplastic activity, (ii) review published preclinical and clinical studies that support anti-colorectal cancer activity and (iii) summarise current clinical trials investigating the potential therapeutic role(s) of u-3 PUFAs at different stages of colorectal carcinogenesis, from adenoma (polyp) prevention to treatment of established malignant disease and prevention of cancer recurrence. INTRODUCTION There is growing epidemiological, experimental and clinical evidence that omega (u)-3 polyunsaturated fatty acids (PUFAs) have anti-colorectal cancer (CRC) activity. u-3 PUFAs may feasibly play a role in several stages of CRC management, from the primary CRC prevention, through to tertiary prevention after treatment of CRC and advanced metastatic disease. In this article, we summarise current knowledge of the mechanistic basis by which u-3 PUFAs are thought to attenuate CRC, before reviewing the experimental data supporting u-3 PUFA supple- mentation for prevention and treatment of CRC. Finally, we review recent clinical data, which now support anti-CRC activity in humans, and summarise current clinical trials of the potential therapeutic roles of u-3 PUFAs at different stages of colorectal carcinogenesis. POLYUNSATURATED FATTY ACIDS Fatty acids are carbon chains with a methyl group at one end and a carboxyl group at the other. Saturated fatty acids contain only carbonecarbon single bonds, whereas unsaturated fatty acids contain one (monounsaturated) or more (poly- unsaturated) carbonecarbon double bonds. Dietary PUFAs of interest are the u-3 and u-6 PUFAs, so named by the position of the rst double bond at the third and sixth carbon from the methyl (u) end respectively (gure 1). Essentialfatty acids are those which are required for biological processes, but which humans are unable to synthesise and must therefore obtain from dietary sources. The parent u-3 PUFA a-linolenic acid and parent u-6 PUFA linoleic acid Significance of this study What is already known about this subject? < The two main omega (u)-3 polyunsaturated fatty acids (PUFAs) found naturally in fish are eicosapentaenoic acid (EPA) and docosahexae- noic acid. < There is strong evidence that u-3 PUFAs have anti-colorectal cancer (CRC) activity from preclinical studies. < Epidemiological evidence links dietary u-3 PUFA intake and reduced CRC risk. < Treatment with u-3 PUFAs is associated with a reduction in measures of mucosal epithelial cell proliferation, a putative biomarker of future CRC risk. What are the new findings? < EPA has chemopreventative efficacy in patients with familial adenomatous polyposis. < u-3 PUFAs may improve the efficacy and tolerability of cancer chemotherapy drugs. < Adjuvant treatment with docosahexaenoic acid may improve breast cancer survival and/or reduce cancer-related cachexia. < The mechanisms of action of u-3 PUFAs probably include reduction of prostaglandin E 2 synthesis and/or synthesis of anti-inflammatory resolvins. How might it impact on clinical practice in the foreseeable future? < An excellent safety and tolerability profile, combined with broader health benefits, make u-3 PUFAs strong candidate cancer chemo- prevention agents. < A role for EPA in ‘sporadic’ colorectal adenoma prevention is currently being evaluated in addition to a potential role in familial adenoma- tous polyposis patients after colectomy. < A phase II trial is currently evaluating the safety and efficacy of EPA in patients with CRC liver metastases. < u-3 PUFAs may find a role for ‘tertiary’ prevention of CRC recurrence or as adjunctive chemotherapy for established CRC. 1 Leeds Institute of Molecular Medicine, University of Leeds, Leeds, UK 2 Department of Hepatobiliary Surgery, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK Correspondence to Professor Mark Hull, Leeds Institute of Molecular Medicine, Wellcome Trust Brenner Building, St James’ University Hospital, Beckett Street, Leeds, LS9 7TF, UK; [email protected] Search strategy and selection criteria: Data for this review were identified by searches of Medline, PubMed and relevant articles using the MESH search terms ‘fish oils’, ‘fatty acids’, ‘colorectal neoplasms’ and ‘liver neoplasms’. Only papers published in English, or including an English abstract, between 1975 and 2010 were included. Ongoing clinical trials were identified by searches of publicly available Clinical Trials Registries (including http://ClinicalTrials.gov/, Current Controlled Trials, WHO International Clinical Trials Registry Platform, National Cancer Institute and NIHR Clinical Research Network Portfolio) and pharmaceutical company registries (including the International Federation of Pharmaceutical Manufacturers and Associations Clinical Trials Portal, Pfizer Clinical Trials and GlaxoSmithKline Clinical Trials Register). Published Online First 13 April 2011 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 135 Recent advances in basic science on July 14, 2020 by guest. Protected by copyright. http://gut.bmj.com/ Gut: first published as 10.1136/gut.2010.233718 on 13 April 2011. Downloaded from
Transcript
Page 1: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

Omega-3 polyunsaturated fattyacids for the treatment andprevention of colorectal cancerA J Cockbain,1 G J Toogood,2 M A Hull1

ABSTRACTOmega (u)-3 polyunsaturated fatty acids (PUFAs) arenaturally occurring substances that are well toleratedand have been used extensively for the prevention ofcardiovascular disease. More recently, u-3 PUFAs havebeen recognised to have anticancer activity. There is alsoevidence suggesting improved efficacy and/or tolerabilityof conventional cancer chemotherapy when administeredwith u-3 PUFAs. The purpose of this review is to(i) describe the mechanisms by which u-3 PUFAs arethought to have antineoplastic activity, (ii) reviewpublished preclinical and clinical studies that supportanti-colorectal cancer activity and (iii) summarise currentclinical trials investigating the potential therapeuticrole(s) of u-3 PUFAs at different stages of colorectalcarcinogenesis, from adenoma (polyp) prevention totreatment of established malignant disease andprevention of cancer recurrence.

INTRODUCTIONThere is growing epidemiological, experimental andclinical evidence that omega (u)-3 polyunsaturatedfatty acids (PUFAs) have anti-colorectal cancer(CRC) activity. u-3 PUFAs may feasibly play a rolein several stages of CRC management, from theprimary CRC prevention, through to ‘tertiary ’prevention after treatment of CRC and advancedmetastatic disease.In this article, we summarise current knowledge

of the mechanistic basis by which u-3 PUFAs arethought to attenuate CRC, before reviewing theexperimental data supporting u-3 PUFA supple-mentation for prevention and treatment of CRC.Finally, we review recent clinical data, which nowsupport anti-CRC activity in humans, andsummarise current clinical trials of the potentialtherapeutic roles of u-3 PUFAs at different stages ofcolorectal carcinogenesis.

POLYUNSATURATED FATTY ACIDSFatty acids are carbon chains with a methyl groupat one end and a carboxyl group at the other.Saturated fatty acids contain only carbonecarbonsingle bonds, whereas unsaturated fatty acidscontain one (monounsaturated) or more (poly-unsaturated) carbonecarbon double bonds. DietaryPUFAs of interest are the u-3 and u-6 PUFAs, sonamed by the position of the first double bond atthe third and sixth carbon from the methyl (u) endrespectively (figure 1).

‘Essential’ fatty acids are those which arerequired for biological processes, but which humansare unable to synthesise and must therefore obtainfrom dietary sources. The parent u-3 PUFAa-linolenic acid and parent u-6 PUFA linoleic acid

Significance of this study

What is already known about this subject?< The two main omega (u)-3 polyunsaturated

fatty acids (PUFAs) found naturally in fish areeicosapentaenoic acid (EPA) and docosahexae-noic acid.

< There is strong evidence that u-3 PUFAs haveanti-colorectal cancer (CRC) activity frompreclinical studies.

< Epidemiological evidence links dietary u-3PUFA intake and reduced CRC risk.

< Treatment with u-3 PUFAs is associated witha reduction in measures of mucosal epithelialcell proliferation, a putative biomarker of futureCRC risk.

What are the new findings?< EPA has chemopreventative efficacy in patients

with familial adenomatous polyposis.< u-3 PUFAs may improve the efficacy and

tolerability of cancer chemotherapy drugs.< Adjuvant treatment with docosahexaenoic acid

may improve breast cancer survival and/orreduce cancer-related cachexia.

< The mechanisms of action of u-3 PUFAs probablyinclude reduction of prostaglandin E2 synthesisand/or synthesis of anti-inflammatory resolvins.

How might it impact on clinical practice in theforeseeable future?< An excellent safety and tolerability profile,

combined with broader health benefits, makeu-3 PUFAs strong candidate cancer chemo-prevention agents.

< A role for EPA in ‘sporadic’ colorectal adenomaprevention is currently being evaluated inaddition to a potential role in familial adenoma-tous polyposis patients after colectomy.

< A phase II trial is currently evaluating the safetyand efficacy of EPA in patients with CRC livermetastases.

< u-3 PUFAs may find a role for ‘tertiary’prevention of CRC recurrence or as adjunctivechemotherapy for established CRC.

1Leeds Institute of MolecularMedicine, University of Leeds,Leeds, UK2Department of HepatobiliarySurgery, St James’s UniversityHospital, Leeds TeachingHospitals NHS Trust, Leeds, UK

Correspondence toProfessor Mark Hull, LeedsInstitute of Molecular Medicine,Wellcome Trust BrennerBuilding, St James’ UniversityHospital, Beckett Street, Leeds,LS9 7TF, UK;[email protected]

Search strategy and selectioncriteria: Data for this reviewwere identified by searches ofMedline, PubMed and relevantarticles using the MESH searchterms ‘fish oils’, ‘fatty acids’,‘colorectal neoplasms’ and ‘liverneoplasms’. Only paperspublished in English, or includingan English abstract, between1975 and 2010 were included.Ongoing clinical trials wereidentified by searches of publiclyavailable Clinical TrialsRegistries (includinghttp://ClinicalTrials.gov/, CurrentControlled Trials, WHOInternational Clinical TrialsRegistry Platform, NationalCancer Institute and NIHRClinical Research NetworkPortfolio) and pharmaceuticalcompany registries (includingthe International Federation ofPharmaceutical Manufacturersand Associations Clinical TrialsPortal, Pfizer Clinical Trials andGlaxoSmithKline Clinical TrialsRegister).

Published Online First13 April 2011

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 135

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 2: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

are both found in vegetable oils. Humans can easilymetabolise linoleic acid to form the u-6 PUFAarachidonic acid (AA). However, endogenousproduction of the u-3 PUFAs eicosapentaenoicacid (EPA) and docosahexaenoic acid (DHA) froma-linolenic acid by humans is so small as to beinsignificant.1 Therefore, the main u-3 PUFAs EPAand DHA are also considered ‘essential’ and areobtained predominantly from cold water, oily fishsuch as mackerel and salmon.PUFAs are biologically important, with roles in

phospholipid membrane structure and function, aswell as cellular signalling and lipid metabolism.PUFAs can be liberated from phospholipidmembranes by the phospholipase A2 family ofenzymes and are metabolised by three main path-ways: (i) the cyclo-oxygenase (COX) pathway, (ii)the lipoxygenase (LOX) pathway and (iii) thecytochrome P450 monoxygenase pathway. Metab-olites derived from the u-6 PUFA AA, such asprostaglandin (PG) E2, are typically proin-flammatory and have been linked with initiationand progression of colorectal carcinogenesis,whereas those derived from u-3 PUFAs (eg, PGE3)are less proinflammatory, and may even have anti-cancer properties. Excellent reviews are alreadyavailable on the metabolism of u-3 PUFAs,2 theimplications of inhibition of AA metabolism on cellproliferation3 and the effects of u-6 and u-3 PUFAmetabolites on colorectal carcinogenesis.4

In addition to their potential role in the treat-ment and prevention of CRC, u-3 PUFAs arebelieved to have a number of other health benefits.u-3 PUFAs (usually a mix of EPA and DHA as fishoil) are widely available in a ‘nutraceutical’ formu-lation. Evidence is strongest for cardiovascularbenefits of the u-3 PUFAs.5 There is some evidenceof benefit in patients who have had a myocardialinfarction5 6 but also uncertainty about whetheru-3 PUFAs have anti- or pro-arrhythmogenicproperties.5 6 u-3 PUFAs have also been variously

implicated in treatment of inflammatory boweldisease, rheumatoid arthritis, age-related maculopathyand neurodegenerative diseases.7

u-3 PUFAs< are ‘essential’ nutrients< are found in large quantities in oily fish< are also widely available as ‘over-the-counter ’nutraceuticals< have broad health benefits, including cardiovas-cular prophylaxis< may have anticancer properties.

MECHANISMS OF THE ANTINEOPLASTIC ACTIVITYOF u-3 PUFASCurrent knowledge of the antineoplastic activity ofu-3 PUFAs has been comprehensively reviewedelsewhere.2 4 8 A brief summary concentrating onmore recent findings is provided here. The threemain antineoplastic activities of u-3 PUFAs thathave been proposed are (i) modulation of COXactivity; (ii) alteration of membrane dynamics andcell surface receptor function and (iii) increasedcellular oxidative stress. Recently described novelanti-inflammatory lipid mediators derived from EPAand DHA, including resolvins, protectins andmaresins, represent a fourth potential antineoplasticmechanism of action of u-3 PUFAs.

Modulation of COX activityCOX-2-dependent synthesis of PGs in colorectalmucosa, in particular PGE2, is believed to playa critical role in the early stages of colorectalcarcinogenesis.9 EPA can act as an alternativesubstrate for COX-2, instead of AA, leading toa reduction in formation of pro-tumorigenic‘2-series’ PGs (eg, PGE2) in favour of ‘three-series’PGs (eg, PGE3) in several cell types including CRCcells (figure 2).2 10 11 PGE3 has anti-tumorigenicactivity against human lung cancer cells in vitro11

and inhibits pro-tumorigenic PGE2-EP4 receptorsignalling in human CRC cells.10 Recently, a ‘PGE2to PGE3 switch’ has been demonstrated in colo-rectal mucosa of rats treated with fish oil.12

However, reduction of PGE2 synthesis and/orgeneration of PGE3 following EPA treatmentremains to be demonstrated in human CRC tissue.It is known that DHA also binds the substrate

channel of COX-2 and inhibits COX-2 activity,13

although the biochemistry of DHA metabolism bythe COX isoforms and the subsequent productionof downstream lipid mediators is poorly understoodcompared with EPA.2

Alteration of membrane dynamics and cell surfacereceptor functionThere is some evidence that the incorporation ofu-3 PUFAs into cell phospholipid membranes altersthe fluidity, structure and/or function of lipid raftsor calveolae.14 These are sphingolipid- and choles-terol-rich microdomains that float freely in the cellmembrane. The localisation of cell surface recep-tors, such as epidermal growth factor receptor

Figure 1 Structure and nomenclature of polyunsaturatedfatty acids (PUFAs). Cx:y u-z refers to the chemicalstructure where x is the number of carbon atoms, y is thenumber of carbonecarbon double bonds and z is theposition of the first carbonecarbon double bond awayfrom the methyl (u) end of the hydrocarbon chain. Notethat all double bonds are in the cis configuration. Smallquantities of conjugated u-3 PUFAs (which containalternating single and double carbonecarbon bonds) alsoexist naturally.

136 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 3: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

(EGFR),15 in lipid rafts is believed to be crucialfor downstream receptor signalling, controllingproliferation and apoptosis.16 17

Increased oxidative stressu-3 PUFAs may have an antineoplastic effectthrough alteration in the cellular redox state andincreased oxidative stress. PUFAs are highly perox-idisable, which generates reactive oxygen species(ROS) such as the superoxide radical. Many tumourcells display altered cellular pathways for thehandling of ROS, including depletion of the majorintracellular antioxidant, glutathione. A subsequentelevation of intracellular ROS levels by u-3 PUFAshas been hypothesised to induce cancer cellapoptosis.18 A potential beneficial interactionbetween u-3 PUFAs and dietary fibre leading toinduction of colonocyte apoptosis has beenelegantly studied by Chapkin and colleagues.19e21

DHA has been shown to potentiate oxidative stressand colonocyte apoptosis induced by the fermenta-tion product short-chain fatty acid butyrate via bothintrinsic and extrinsic apoptosis pathways.22e24

The relative contributions of, and interactionsbetween, these three activities to the anticancerproperties of u-3 PUFAs are unclear. For example,u-3 PUFAs might alter EGFR function by changingreceptor behaviour in lipid rafts but EPA might alsodecrease trans-activation of EGFR by reduction inPGE2 synthesis.25 Activation of peroxisome prolif-erator-activated receptors by u-3 PUFAs is alsorecognised but it is not known whether this occursdirectly or via changes in COX metabolism, therebyaltering levels of PGD2 or 15-deoxy PGJ2.26

Novel anti-inflammatory lipid mediatorsIn the presence of aspirin, which irreversibly acet-ylates the COX enzyme, EPA drives COX-2-depen-dent production of resolvin (Rv) E1 (5S,12R,18R-trihydroxyeicosapentaenoic acid) via metabolism of18R-hydroxyeicosapentaenoic acid by 5-LOX (figure2).27 18R-RvE1 has been detected in plasma ofhealthy volunteers in ng/ml quantities after aspirinand EPA ingestion.28 More recently, synthesis of the18S enantiomer of RvE1 has been demonstratedafter EPA and aspirin treatment in healthy volun-teers.29 The precursors of E-series resolvins may alsobe produced independently of COX by directcytochrome P450 metabolism of EPA.30

Metabolism of DHA can produce D-seriesresolvins via a LOX-dependent pathway to produce17S-resolvins, or via acetylated-COX-2 leading to17R-resolvin synthesis.31 DHA can also be metab-olised by leucocyte-mediated pathways to produce17S-docosatrienes termed protectins31 or bymacrophage-mediated pathways to produce14-LOX-derived products termed maresins.32 Thesenewly described families of EPA- and DHA-derivedlipid mediators all share anti-inflammatory andinflammation resolution activity in animal modelsof acute inflammation.32 33

Cell signalling via these novel lipid mediatorfamilies is best characterised for RvE1. Both 18Rand 18S enantiomers of RvE1 are ligands forChemR23 and BLT1 G protein-coupled receptors.29

RvE1 has been shown to induce expression ofintestinal alkaline phosphatase in human CRC cellsin a ChemR23-dependent manner and abrogatechemically induced colitis in mice.34 It is currentlynot known whether u-3 PUFA-derived resolvinsexhibit antineoplastic activity. However, it isknown that ChemR23-dependent RvE1 signallinginhibits nuclear factor kB activation, which isa critical regulator of early-stage colorectalcarcinogenesis.35

Although u-3 PUFAs have recognised immuno-modulatory activity, including alteration of T-cellactivation and cytokine production,36 the contri-bution of any effect on host antitumour immunesurveillance to the anticancer activity of u-3 PUFAshas received relatively little attention, to date. Invitro studies have suggested that both EPA andDHA may suppress angiogenesis.37 38 Whethernegative regulation of stromal-epithelial cellsignalling and/or the angiogenic response contrib-utes to the antineoplastic activity of u-3 PUFAs invivo requires further investigation.

Figure 2 The effect of eicosapentaenoic acid (EPA) without or with concurrent aspirintreatment on cyclo-oxygenase (COX) activity. (A) When arachidonic acid (AA), derivedmainly from dietary u-6 polyunsaturated fatty acid (PUFA) linoleic acid in ‘westernised’diets, is the main substrate for COX-1 and COX-2, the predominant prostaglandin (PG)species in colorectal tissue is PGE2. COX-2 is the predominant COX isoform in dysplasticand malignant colorectal tissue.9 (B) EPA can act as an alternative substrate for bothCOX enzymes. It effectively inhibits COX-1 activity but modulates the enzymatic activityof COX-2 so that PGH3 is the major product.

2 PGH3 is then converted to other three-seriesPGs such as PGE3 by downstream PG synthases. Although the KM for EPA is similar toAA, the enzymatic turnover for EPA is approximately threefold less than that of AA sothat the overall outcome is reduction in COX-2-dependent PGE2 synthesis andsimultaneous appearance of smaller quantities of PGE3.

2 (C) Aspirin irreversiblyacetylates COX-1 and COX-2. This leads to effective inhibition of COX-1 activity.However, acetylated COX-2 can metabolise EPA producing 18R-hydroxyeicosapentae-noic acid (HEPE) and 18S-HEPE instead of PGH3. In turn, HEPEs can be metabolised by5-lipoxygenase (5-LOX) generating E-series resolvins, in combination with a reduction intissue PGE2 levels.

28 29 D-series resolvins are generated via docosahexaenoic acidmetabolism by lipoxygenase-dependent pathways.

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 137

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 4: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

u-3 PUFAS FOR THE PREVENTION OF CRCEpidemiological observationsA link between dietary u-3:u-6 PUFA balance andCRC risk originated from epidemiological studiesthat observed reduced rates of CRC in Greenlandand the Far East compared with Western popula-tions.39 While the results of epidemiological studieshave been variable, they have tended, in general, toreport a small reduction in CRC risk withincreasing dietary fish intake, a view supported bythe second expert report into food, nutrition andthe prevention of cancer by the World CancerResearch Fund and the American Institute forCancer Research in 2007.40

Interpretation of epidemiological studies hasbeen hampered by heterogeneity in study design.The use of food questionnaires to record dietaryintake is subjective, and does not always discrimi-nate between oily fish such as sardines (high in u-3PUFAs) and lean fish such as cod (lower u-3 PUFAcontent). Moreover, studies do not alwaysdiscriminate between processed (smoked or salted)and non-processed fish, which may confoundobservational data on CRC risk.40 A meta-analysisof 19 prospective cohort studies was limited bystudy heterogeneity and wide variability in bothfrequency of fish intake and the reporting ofoutcome measures (eg, grams/day, portions/week,u-3 PUFA g/day).41 Meta-analysis of CRC inci-dence data could be performed on only 14 of 19studies. It demonstrated a small but significant 12%relative risk (RR) reduction in CRC incidencebetween the highest (1e7 portions or 210e590 gfish per week) and lowest (0e1 portions or 0e100 gfish per week) fish consumption groups. The pooledRR reduction rose to 22% in those studies wherethe difference in fish consumption between thehighest and lowest categories was at least sevenportions of fish a month.Since this meta-analysis, two large observational

studies have demonstrated significant inverse rela-tionships between u-3 PUFA intake/levels and riskof colorectal neoplasia. The first, a caseecontrolstudy of 1872 patients (929 cases of distal CRC and943 controls), estimated u-3 and u-6 PUFA intakefrom detailed one-to-one dietary interviews.42 Thestudy showed a significant dose-dependent reduc-tion in CRC risk for total u-3 PUFA intake(OR¼0.61 for the highest vs lowest quartile), aswell as for EPA and DHA intake individually. In thesecond study, serum PUFA levels were measured in861 patients undergoing endoscopy.43 Threehundred and sixty-three patients were diagnosedwith a colorectal adenoma(s) and 498 polyp-freepatients served as controls. When comparing serumu-3 PUFA levels (as a percentage of overall PUFAlevels), there was a significant reduction in colo-rectal adenoma risk (OR¼0.67) between the firsttertile (<1.8% u-3 PUFA) and the third tertile(>2.3% u-3 PUFA). By contrast, there wasa significant increase in colorectal adenoma risk(OR¼1.68) between the first tertile (<58.9%) andthird tertile (>62.8%) of serum u-6 PUFA content.

Animal studiesThe differential effect of u-3 and u-6 PUFAs hasbeen demonstrated in a number of animal modelsof early-stage colorectal carcinogenesis relevant toprevention of CRC. These studies are summarisedin table 1. Injection of the carcinogens azoxy-methane or 1,2-dimethylhydrazine drives develop-ment of colorectal aberrant crypt foci (ACF) after6e8 weeks, before development of colorectaladenomas and adenocarcinomas from 8 monthsonwards. These lesions have all been widely used asend points for comparison of the effects of u-3 andu-6 PUFA supplementation on ‘sporadic’ colorectalcarcinogenesis. Alternatively, the effect of u-3 andu-6 PUFAs on adenoma number in the ApcMin/+

mouse model of familial adenomatous polyposis(FAP) has been investigated.Studies of rodents fed an u-3 PUFA-supple-

mented diet versus an equivalent u-6 PUFA-supplemented diet or low-fat control diet controls(table 1) have consistently reported a 20e50%reduction in chemically induced tumour inci-dence,19 45 47 48 50 52 53 55 56 64 together witha 30e70% reduction in tumour multiplicity,in both carcinogen and ApcMin/+ mousestudies.44 45 50 53 58 59 61 64 Studies using thenumber of ACFs as the primary end point (table 1)have reported an effect of similar magnitude withu-3 PUFA supplementation.54e56 59 Reddy andSugie demonstrated that u-3 PUFA supplementa-tion reduced chemically induced colonic tumourincidence when given in either the pre- or post-initiation phase compared with u-6 PUFA supple-mentation.46 When given in both phases, tumourincidence was further reduced to half that seen inlow-fat control animals. Further studies havedemonstrated that these effects are directly relatedto u-3 PUFA supplementation rather than simplya reduction in u-6 PUFA intake.53 65 66 While mostin vivo studies have compared a mixture of EPAand DHA, attention is drawn to the few studieseither directly comparing EPA and DHA,60 or usingEPA45 61 64 or DHA49 51 58 as single agents. Ingeneral, similar results have been demonstratedwith each of the two main u-3 PUFAs. The effectof fish oil administration on azoxymethane-induced ACF multiplicity in rats has also beentested in combination with the fibre fermentationproduct butyrate. Crim and colleagues havedemonstrated that butyrate decreases ACF numberin rats administered fish oil, but not corn oil (rich inu-6 PUFAs).20

One recent study demonstrated that dietarysupplementation with a large amount (6%) of DHAexacerbated colonic inflammation and dysplasia ina mouse model of infection-induced colitis-associ-ated cancer.57 The doseeresponse relationshipbetween u-3 PUFA intake and intestinal tumori-genesis in animal models of colitis-associated cancerrequires further investigation.Analysis of mucosal PUFA content has consis-

tently demonstrated incorporation of u-3 PUFA,at the expense of AA content, in rodents

138 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 5: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

Table1

Invivo

pre-clinicalstudiesof

u-3

PUFA

supplementationforthepreventionof

CRC

Study

Model

NTreatm

entgroups

Timing/duration

Outcomemeasure(s)

Results

(maximal

changesu-3

PUFA

groupvs

Ctrlunless

stated)

Carcinogen-inducedmodels

Nelsonet

al44

Sprague-Daw

leyrat

DMH

5017%MO1vs

17%COvs

Ctrl

7wkpre-

and17

wkpost-DMH

Tumourincidenceandmultiplicity

Incidenceof

metastases

4Tumourincidence,

35%Y

tumourmultiplicity,

4incidenceof

metastases

Minoura

etal45

Donryurat

AOM

100

4.7%

EPAvs

5%LA

15wkpre-

and20

wkpost-AOM

Tumourincidence

TumourandcolonicmucosalPU

FA+

PGE 2

content

50%YTumourincidence,

75%Ymultiplicity,

[EPA&Y

AAin

tumourandmucosa,

80%YtumouralPG

E 2Reddy

46

F344

rat

AOM

234

4%e12%MO1vs

24%&5%

CO

38wkpost-AOM

Tumourincidence

TumourPU

FAcontent

50%[

Tumourincidence+

multiplicity

in24%

COvs

allothergrps,[

tumouralEPA+DHA

with

MOdiets

Deschneret

al47

CF1

mouse

AOM

300

4e16%Max

EPAvs

4%and

20%CO

2wks

pre-

&1e

45wkpost-AOM

ColonicmucosaPI

&FA

D1wk

Tumourincidence45

wk

DosedependentY

FAD,38%Y

PIin

16%MO

grpvs

20%CO,50%Y

tumourincidence16%/

10%MOgrp

Reddy

48

F344

rat

AOM

273

18%MO2vs

23.5%and5%

CO

2wkpre-

&36

wkpost-AOM.Diet

crossover3/7post-AOM.

Tumourincidence

YTumourincidenceandmultiplicity

whenMO

vs23.5%COgivenineither

initiationor

post-

initiationphases

Takahashiet

al49

F344

rat

DMH

970.7mlDHAvs

0.7mlH2Oig

daily

1daypre-

and4/8/12

wkpost-DMH

No.

ofcolonicACFs

Serum

chol

andPU

FAcontent

60%Y

ACFs

(YACFs

seen

whenDHAgiven

ineither

initiationor

post-initiationphase),

50%Y

AA,50%[

DHAandEPA,20%Y

chol

Hendrickse5

0Wistarrat+

colon

anastomosisvs

sham

AOM

160

20%FO

1vs

20%CO

3wkpre-

and15/23wkpost-AOM

ColonicmucosalPI

Tumourincidence,

size,no.

MucosalPU

FAcontent

40%Y

Tumourincidence,

50%Y

multiplicity,

60%Y

peri-anastamotic

tumours.YPI

[EPA/

DHA&90%Y

AAin

tumour/mucosa

Chang

19

Sprague-Daw

leyrat

AOM

260

11.5%FO

2vs

15%CO6

6%cellulose

vs6%

pectin

1weekpre-

&16/32wkpost-AOM

Colonictumourincidence

Crypt

PI/AI/celldifferentiation

20%Y

Tumourincidence,

4PI,20%[AI,

[celldifferentiation,

[AIFO

-pectin

grpvs

all

otherfat-fibre

grps

Takahashiet

al51

F344

rat

AOM

961mlDHAvs

1mlwater

igdaily

4/12/36wk

No.

ofACFandtumours

PlasmaPU

FAsandPG

E 2

25%Y

ACF(w

k4/12)&35%Y

tumourmultiplicity,

50%Y

plasmaPG

E 2,75%Y

plasmaAA,

303[

EPA,63

[DHA(w

k36)

Goodet

al52

F344

rats

AOM

161

18%MO3vs

5%and23%CO

5%CO12

wkpost-injectionthen

6e12

wkexperim

entaldiet

No.

ACF

No.

+size

colonictumours

15e20%[

ACFvs

both

COgrps,25%Y

tumour

incidencebut50%[

tumoursize

MOvs

COgrps

(bothNS)

Singh

53

F344

rat

AOM

144

21%FO

3vs

24%COvs

Ctrl

1/12/36wkpost-AOM

Tumourincidenceandmultiplicity

FOgrp30%Y

tumourincidenceandmultiplicity.

23.5%COgrp33%[tumourincidenceand90%

[multiplicity

Latham

54

Wistarrat

DMH

688%

FO4vs

8%CO

FOvs

CO24/48hrs

post-DMHthen

18wkCO

Crypt

cellAI&PI

(24/48

h)No.

ACF(18wk)

[AIandY

PIat

24/48h,

50%YACFat

18wk

Rao

etal55

F344

rat

AOM

360

17%FO

3vs

5%COvs

20%

mixed

lipids

CO2wkpre-AOM

Experim

entaldietsfor8e

38wk

post-AOM

No.

ACF,

tumourincidence

ColonicmucosaAI

44%Y

ACFs,30%Y

tumourincidence,

60%

Ytumourmultiplicity

23[AIvs

20%mixed

lipid

diet

Crim

etal20

Sprague-Daw

leyrat

AOM

8011.5%FO

2vs

15%CO6

5%butyrate

3wkpre-

and8wkpost-AOM

No.

ACF,

colonicmucosaAI

[ACFs

CO+

butyrate

vsallgrps,Y

largeACFs

and[AIinFO

+butyrate

grpvs

FOaloneor

control

Vanam

alaet

al12

Sprague-Daw

leyrat

AOM

2015%FO

5+

pectin

vs15%CO

+cellulose

32days

pre-

and31

wkpost-AOM

ColonicmucosaAI

ColonicmucosaPG

E 2/PGE 3

b-cateninandPPARdexpression

23[

AIand78%Y

mucosalPG

E 2,[PG

E 3(PGE 3

notdetected

inCOgroup),Y

b-cateninandPPARd

expression

Moreira

etal56

Wistarrat

DMH

2018%FO

6vs

18%SOYO

2wkpre-

&36

wkpost-DMH

No.

ACFs,adenom

aincidence

Colon/liverPU

FAcontent

47%Y

ACF,

80%Y

adenom

aincidence,

53[

u-3-PUFA

incolon+

liver,60e75%Y

n-6PU

FAin

colon+

liver

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 139

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 6: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

supplemented with u-3 PUFA compared withcontrols,45 49 50 together with a reduction inmucosal PGE2,12 45 60e62 reduction in mucosal cellproliferation47 50 54 and an increase in mucosal cellapoptosis 12 19 20 54 62 (table 1).

u-3 PUFAS IN THE TREATMENT OF CRCIn vitro studiesMany in vitro studies have explored the antineo-plastic activity of u-3 PUFAs against human CRCcells, with both EPA and DHA treatment beingassociated with reduced cellular proliferation67e74

and increased apoptosis.68 69 72 75 It remains unclearfrom the few studies comparing EPA and DHAwhether there is any significant difference in anti-proliferative and/or pro-apoptotic activity. Calvielloet al reported a more pronounced reduction in cellnumber with EPA than with DHA,71 whereas Chenand Istfan have reported a lower cell number withDHA than with EPA, and that only DHA inducedapoptosis.72 Both u-3 PUFAs have been shownto reduce COX-2 expression and PGE2 produc-tion.10 69 71 76 u-3 PUFAs also reduce cell growth ofCOX-negative CRC cells, suggesting that the anti-CRC activity of u 3-PUFA occurs via both COX-dependent and independent mechanisms.67

Increased membrane fluidity73 and lipid perox-idation,73 74 as well as reduced vascular endothelialcell growth factor, b-catenin, peroxisome prolifer-ator-activated receptor g (PPARg), BCL-2, matrixmetalloproteinase levels and reduced extracellularsignal-related kinase-1/2 signalling, have all beendemonstrated in human CRC cell lines treated withu-3 PUFAs.17 69 71 72 75 77

Animal studiesThe effect of u-3 and u-6 PUFA supplementationon the growth of human CRC cell lines grown asxenograft tumours in immunocompromised micehas been studied widely (table 2). There has beena consistent 40e60% reduction in xenograft size inrodents supplemented with u-3 PUFAs comparedwith controls.67 71 81 Similar findings have beenreported for studies of rodent CRC cell allografttumours (table 2).65 78 79 82 84 Similar to data fromchemoprevention models, the u-3 PUFA content oftumours increased, at the expense of AA, in animalssupplemented with u-3 PUFA,67 79 80 82 togetherwith a reduction in expression of COX-2,65 71

reduction in tissue PGE2 levels71 and a reduction intumour vascularity.71 Conjugated EPA (figure 1) hasbeen shown to have antitumour activityagainst DLD-1 human CRC cell tumours in nudemice, which was associated with increased lipidperoxidation.83

Models of CRC metastasisFewer preclinical studies have investigated theeffect of u-3 PUFA supplementation on the devel-opment of metastatic disease. Two studies havemodelled liver metastasis by injection of CRC cellsinto the portal vein or superior mesenteric vein, anda third by injection into the spleen. Iwamoto et alTa

ble1

continued

Study

Model

NTreatm

entgroups

Timing/duration

Outcomemeasure(s)

Results

(maximal

changesu-3

PUFA

groupvs

Ctrlunless

stated)

Woodw

orth

etal57

SMAD3�

/�mouse

Helicobacterinduced

colitis

122

0.75%e6%

DHAvs

6%SAFO

vs7%

COvs

Ctrl

1.8wkpre-infection

2.8wkpre-

and4wkpost-infection

Colon

inflammation/dysplasia

HepaticPU

FAcontent

Bodywt

[Inflammation/dysplasia2.25e6%

DHAvs

Ctrl.

53[

hepatic

DHAcontent,85%less

wtgain

and

10e18%Y4w

ksurvivalin

6%DHAgrpvs

CO/

SAFO

/Ctrl

Apc

mouse

models

Oshimaet

al58

Apc

Δ716

203%

DHAvs

Ctrl

7wk

Colonicpolypno.andsize

69%Y

Polypno.in

femalemiceonly,4

polyp

no.in

malemice,

Ypolypsize,moremarkedin

femalemice

Paulsenet

al59

Apc

Min/+

510.4e

2.5%

FO7vs

12%CO

17wk

No.

ACFs

andadenom

as48e66%Y

no.and26e38%Y

size

oftumours,

YACFs

infemalemiceon

2.5%

diet

only

Petricket

al60

Apc

Min/+

773.1%

EPAvs

3.1%

DHAvs

Ctrl

7wk

Tumoursize

+no.

MucosalPU

FA+

PGE 2

30%/50%

YTumourno.DHA/EPA

grpvs

Ctrl,

15%Ytumoursize

EPA/DHAgrpvs

Ctrl,50%Y

PGE 2

inEPA/DHAgrpvs

Ctrl

Petricket

al61

Apc

Min/+

201.5%

EPAvs

1.5%

AAvs

Ctrl

8wk

Tumoursize

+no.

MucosalPU

FA+

PGE 2

54e68%Y

Tumourno.&18%Y

tumoursize

EPAvs

CtrlandAAgrps.74%Y

mucosalPG

E 2EPAvs

AAgrp

Boseet

al62

Apc

Min/+

9512%MO4vs

20%mixed

lipid

diet

9wk

Tumourno.+

size

TumourPI,AI,PG

E 2,b-catenin

4Tumourno,50%Y

no.tumours

>2cm

,3.73

[AI,4

PI,89%Y

PGE 2,62%Yb-catenin

Finiet

al63

Apc

Min/+

482.5%

and5%

EPAvs

Ctrl

12wk

MucosalPU

FA+

COX-2

Polypno.+

size,body

weight

72%/79%

YPolypsize

(2.5%/5%EPAgrp)

YCOX-2

and[

EPAin

EPAgrps,YweightCtrlgrp

4,no

significant

difference;Y,decrease;[,increase.

AA,arachidonicacid;A

CF,aberrantcryptfoci;A

I,apoptosisindex;AOM,azoxymethane;chol,cholesterol;CO,cornoil;Ctrl,control;CRC,colorectalcancer;DHA,docosahexaenoicacid;D

MH,1,2-dimethylhydrazine;EPA

,eicosapentaenoicacid;FAD,focalarea

ofdysplasia;FO

,fish

oil(FO

18%EPA/15%

DHA;FO2¼

unspecified

u-3

PUFA

content;FO

31%u-3

PUFA

;FO4¼

18%EPA/8%DHA;FO5¼

18%EPA/11%

DHA;FO6¼

24%EPA/20%

DHA;FO

54%EPA/30%

DHA);ig,intragastric;LA,linoleicacid;M

axEPA,18%

EPA+

12%DHA;MO,m

enhadenoil(MO116%EPA+

11%DHA;MO22.4%

EPA+

11%DHA;MO3¼

unspecified

u-3

PUFA

content;MO4¼

13%EPA/12%

DHA);PG

,prostaglandin;PI,proliferationindex;PPAR,peroxisom

eproliferator-activated

receptor;PU

FA,

polyunsaturatedfattyacid;SAFO

,safflow

eroil;SOYO

,soybeanoil;wk,

weeks;wt,weight.

140 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 7: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

demonstrated a 40% reduction in number and 44%reduction in size of liver metastases in ratssupplemented with EPA (9.5% w/w as the ethylester) compared with controls. This was associatedwith a reduction in the tumour cell proliferationindex (PI), but no change in the apoptosis index(AI), and downregulation of vascular cell adhesionmolecule 1 (VCAM-1). Rats fed a diet high in u-6PUFA showed a threefold increase in number and1.5-fold increase in the size of metastases.85 A morerecent study of an EPA/DHA mixture (1.96% fishoil; EPA:DHA ratio 3:2) demonstrated a 70%reduction in incidence and 50% reduction in size ofhepatic and extra-hepatic metastases, as well asa 30% reduction in tumours expressing VCAM-1.86

By contrast, Griffini et al demonstrated a 10-foldincrease in liver metastases in rats supplementedwith an EPA/DHA mixture (20% fish oil v/w; EPA:DHA ratio approximately 3:2) compared with 5%soybean controls, and a fourfold increase comparedwith those animals supplemented with 20%safflower oil (high in u-6 PUFA).87 This was asso-ciated with an increase in liver:body weight ratioand an increase in mitotic tumour cells in the u-3PUFA group. These results are difficult to reconcilewith the other preclinical data, but the 20% fish oilpreparation used in this study is a much higherdose than that used in any other in vivo study, andis far in excess of any clinically attainable dose inhumans.Two further studies have modelled the effect of

u-3 PUFA supplementation on the development ofpulmonary metastases, by measuring lung coloni-sation after injection of CRC cells into the tailvein.78 79 One demonstrated that supplementationof either EPA or DHA (0.1 ml aliquots daily of 98%pure EPA or DHA ethyl ester) was associated withsignificantly fewer lung colonisations at 12 dayscompared with controls (54% and 58% fewercolonies, respectively).79 The other study foundthat while supplementation with high-fatsafflower oil (24.7% by weight for 30 days preino-culation) caused a fivefold increase in the number ofpulmonary colonies, there was no difference in low(5%) or high (24.7%) u-3 PUFA intake on thenumber of metastases compared with controls.78

PHARMACOLOGY, SAFETY AND TOLERABILITY OFu-3 PUFAS IN HUMANSBioavailability of u-3 PUFA preparationsThe bioavailability of u-3 PUFAs is highly variableand is dependent on the form of the u-3 PUFA, themode of administration and the timing of admin-istrationdfor example, with food. Dietary PUFAsare absorbed primarily in the small intestine. u-3PUFAs in the free fatty acid (FFA) form areabsorbed more efficiently than those in thetriglyceride or ethyl ester form.88 89 Lawson andHughes demonstrated >95% absorption of an oraldose of EPA and DHA (as measured by plasmaPUFA content) when given to healthy volunteers inthe FFA form, whereas absorption in the triglyc-eride and ethyl ester forms was approximately 65%Ta

ble2

Invivo

pre-clinicalstudiesof

u-3

PUFA

supplementationforthetreatm

entof

CRC

Study

Model

NTreatm

entgroups

Timing/duration

Outcomemeasure(s)

Results

(changes

areforu-3

PUFA

groupvs

controlgroupunless

stated)

Cannizzo

78

BALB/c

mouse,CT26CRC

cells

into

1)descending

colon2)

tailvein

330

5%and25%MO1vs

5%and25%SAFO

30days

pre-

and28

days

post-injection

1.Tumoursize

2.No.

lung

mets

3e53

YTumoursize

MOvs

SAFO

grps,[

lung

metsin

25%SAFO

grpvs

all

othergrps

Iigoet

al79

CDF1

mouse

Co26Lu

CRCcells

subcut

230

0.1/0.2mlEPAvs

DHAvs

OAvs

LAig

daily

Day

5e21

post-injection

Tumoursize,no.lung

mets

Plasma/tumourPU

FAEPA/DHAgrps

dose

dependentYtumoursize

andYlung

mets,

[EPA/DHAandYAA

inplasmaandtumour

Calderet

al80

Nudemouse

HT-29

CRCcells

subcut

9020%CO/SAFO

/MO2/OO

vs2.5%

COCtrl

3wkpre-

and2wk

post-injection

Tumoursize

andPU

FAcontent

65e74%[Tumoursize

inhigh-fat

grps

except

MO,[tumourEPA/DHA+

57%YAA

inMOgrp

Boudreauet

al67

Nudemouse

HCT116

CRCcells

subcut

102

18%MO3vs

20%SAFO

2wkpre-

and3wk

post-injection

Tumoursize

TumourPU

FAcontent

50%Y

Tumoursize,[

tumourEPAand50%Y

tumourAA

Kato

etal81

Nudemouse

WiDRCRCcells

subcut

248%

and24%CO16%MO4

vs16%GAO

53days

post-injection

Tumourwt

Tumourwtvs

8%COtumourwt:,87%Y

(GAO),54%Y

(MO),36%[

(24%

CO)

Togniet

al82

Wistarrat

Walker256tumourcells

subcut

781g/kg

body

wt.MaxEPA

vsCOvs

Ctrl

10wkpre-

and2wk

post-injection

TumourwtandPU

FAcontent

Cachexiabiom

arkers

(serum

glucose,

chol,lactate,

andliver/m

uscleglycogen)

40/60%

YTumourwtvs

CO/Ctrl,63/42%

YtumourAA:EPA

vsCO/Ctrl,Ycachexia

biom

arkers

Calviello

etal71

Nudemouse

HT-29

CRCcells

subcut

451g/kg

body

wtEPAvs

DHAvs

H2OCtrlig

1wkpre-

and4wk

post-injection

Tumoursize,AI,PI,MVD

TumourCOX-2

expression

TumourPG

E 2

Nodifference

ineffect

EPAvs

DHA,EPA/DHAvs

Ctrlboth

show

ed55%Y,tumour

size,50%Y

PI,1.5x[

AI,w

50%Y

MVD,40%Y

PGE 2,30%YCOX2andYVEG

F

Tsuzuki83

Nudemouse

DLD-1

CRCcells

subcut

4050

mg20%CLA

/EPA

/CEPA

daily

vs50

mgSAFO

Ctrl

4wkpost-injection

Tumourwt,DNAfragmentation

Mem

branephospholipid

peroxidation

TumourwtY80%CEPAvs

Ctrl.,

DNAfragmentation[43

CEPAvs

Ctrl,

[phospholipid

peroxidationCEPAvs

allgrps

Pizato

etal84

Wistarrat

Walker256tumourcells

subcut

6020%FO

1vs

20%SUNO

vsCtrl

8wkpre-

and2wk

post-injection

Tumourwt

Lipidperoxidationproducts

Cachexiabiom

arkers

(blood

glucose,

TAG,chol)

60%YTumourwt,34%[

lipid

peroxidationproducts,Ycachexiabiom

arkers

Mundet

al65

Wistarrat

Walker256tumourcells

subcut

701g/kg

FO2vs

COvs

Ctrl

70days

pre-

and14

days

post-injection

Tumourweight,AIandCOX-2

Lipidperoxidationproducts

PlasmaPG

E 2

Tumourweight50%Y(FO)and30%[(CO),AI4x

[(FO)and50%Y

(CO),COX-2

45%Y(FO)and50%[(CO),PlasmaPG

E 2Y

(FO)and4

(CO),FO

33[

lipid

peroxidationproducts

[,increase;Y,decrease;4

,no

significant

difference.

AA,arachidonicacid;A

I,apoptosisindex;chol,cholesterol;CO,cornoil;CRC,colorectalcancer;Ctrl,control;DHA,docosahexaenoicacid;EPA

,eicosapentaenoicacid;FO,fish

oil(FO

13%EPA/20%

DHA,FO

18%EPA/12%

DHA);GAO,goldenalgaeoil(DHA

only);grp,group;grps,groups;ig,intragastric;LA,linoleicacid;M

axEPA,18%

EPA/12%

DHA;m

ets,metastases;MO,m

enhadenoil(MO1,M

O3,M

O4u-3PU

FAcontentunspecified,M

O211%EPA+5.2%

DHA);MVD,m

icrovesseldensity;O

A,oleicacid;O

O,oliveoil;

PI,proliferationindex;SAFO

,safflower

oil;subcut,subcutaneous;CLA

,conjugatedlinoleicacid;CEPA,conjugatedEPA;P

G,prostaglandin;PU

FA,polyunsaturated

fattyacid;S

UNO,sunflower

oil;TA

G,triacylglycerol;VEG

F,vascular

endothelialgrowth

factor;w

t,weight.

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 141

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 8: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

and 21%, respectively.89 These differences arebelieved to be due to the ability of enterocytes todirectly absorb u-3 PUFAs in the FFA form,whereas absorption of those in the ethyl ester andtriglyceride forms requires prior hydrolysis bypancreatic lipase.89 Pancreatic lipase hydrolysesfatty acids from the three positions of the triglyc-eride backbone at different rates.90 Bioavailability istherefore further complicated by variation in thedistribution of fatty acids in the sn-1, sn-2 and sn-3positions in fish oils from marine mammals, marinefish and processed fish oil supplements.91 92 Co-absorption with other dietary fats, absorption asmicelles rather than direct absorption through theintestinal wall and the variable action of gastriclipase and gastric absorption can also affectbioavailability. These variables make it difficult toobtain direct comparisons between dose andoutcomes in different clinical (and animal) studies.

Duration of u-3 PUFA supplementationThere is considerable variation in the duration ofu-3 PUFA supplementation in clinical studies,ranging from 4 weeks to 6 months. These aresummarised in table 3. Studies have demonstratedthat maximal incorporation of orally administeredu-3 PUFA into plasma and colorectal mucosaoccurs within 3 weeks.103 104 Hillier et al gave fishoil (3.2 g EPA+2.2 g DHA daily) to 11 patients withactive inflammatory bowel disease and took colonicbiopsy specimens at 3, 6 and 12 weeks.104 After3 weeks, mucosal EPA content had increasedsevenfold and DHA content had doubled. Theselevels were maintained but did not increase furtherthroughout the 12 weeks’ study period. Bycontrast, AA content did not fall significantly untilafter 6 weeks of treatment. Colonic mucosal PGE2content fell to 45% of baseline values after 3 weeksof fish oil supplementation, and fell only slightlyfurther to 43% of the baseline value at 12 weeks.Similarly, in eight healthy volunteers taking1.4e4.2 g daily EPA for 12 weeks, a significant,maximal increase in EPA content of plasma, plate-lets, neutrophils, monocytes and lymphocytes wasobserved at 2 weeks, whereas AA content fellsignificantly only by week 12.103

These results support the hypothesis that theincreased mucosal EPA and DHA content, ratherthan the reduction in u-6 PUFA content, contrib-utes to the reduction in PGE2 levels. Tissue u-3PUFA incorporation in humans plateaus after a fewweeks. The optimal duration of u-3 PUFA treat-ment required to produce maximal antineoplasticeffects remains to be established.

Adverse effectsu-3 PUFA formulations are generally very welltolerated. Table 3 summarises the adverse effectsreported in human studies of the effect of u-3PUFA supplementation on colorectal mucosalparameters and colorectal polyps. Study with-drawal due to EPA intolerance in a recent phase IIItrial of EPA-FFA 2 g daily was only 3.4%.102 Ina previous phase II trial with the same preparation,Ta

ble3

Clinicalstudiesof

u-3

PUFA

treatm

enton

colorectalmucosabiom

arkers

andpolypnumber

Study

Design

Nu-3

PUFA

dose

Treatm

entduration

Primaryoutcom

eTissue

PUFA

content

Sideeffects(athighestgivendose)

Results

Mucosalbiom

arkerstudies

Antiet

al93

R,DB,PC

‘sporadic’adenom

a24

7.7gFO

1daily

12wk

PI[EPAandYAA

Not

reported.Nodropouts

inFO

grp

62%YPI

Bartoliet

al94

R,DB,PC

‘sporadic’adenom

a40

2.5e

7.7gFO

1daily

30days

PIDosedependent[

EPA/DHA&YAA

Not

reported.

Dosedependent

40e70%Y

PI

Bartram

etal95

DBcrossovertrial

Healthyvolunteer

124.4gFO

2daily

4wk+4wk

PIu-3

PUFA

4u-6

PUFA

Y(NS)

Mild

fishodour(9/12)

16%Y

PIand35%Y

mucosalPG

E 2Antiet

al96

R,DB,PC

‘sporadic’adenom

a60

2.5e

7.7gFO

1daily

30days

PIDosedependent[

EPA/DHAandYAA

2/15

Dropout,5/15

fishodour,

1/7diarrhoea

Doseindependent

50e70%YPI

Huang

etal97

R,DB,PC

Dukes

A/B

CRCor

severely

dysplastic

polyp

277.2gFO

3daily

6months

PI[EPA/DHA&YAA

NoSEs

observed

71%YPI

(onlyin

patients

with

high

baselinePI)

Gee

etal98

R,PC

,singleblind

AwaitingCRCsurgery

512.4gFO

4daily

7-21

days

pre-

and

8e12

wkpost-surgery

PI[EPA/DHA,

[u-3:u-6

ratio

1Dropout

dueto

capsuleintolerance

(grp

notstated)

Noeffect

onPI

atsurgeryor

12wkpost-op

Cheng

etal99

R,C,open

label

Previous

CRC/adenoma

41Dietary

advice

+-500mg

FO5daily

2years

PI/AI

Not

assessed

Not

reported

PI4

,50%[AI,50%[

Bax,COX-2

4

Courtneyet

al100

R,singleblind

‘sporadic’adenom

a30

EPA2gdaily

asfree

fattyacid

3months

PI/AI

[EPA/DHA&YAA

1/15

Dropout

each

grp

3/15

mild

diarrhoea+

2/15

abdo

pain

20%YPI,73

[AI

Westet

al64

R,DB,PC

‘sporadic’adenom

a152

EPA1g/2gdaily

asfree

fattyacid

6months

PI/AI

[EPA/DHA&YAA

Not

reported

13%YPI,57%[

AI(NS)

Polypend-pointstudies

Akedo

etal101

Open-label

FAP

5Dietary

advice

and2.8g

FO6daily

1e2years,

?trial

stoppedearly

Polypno.

Not

assessed

13Diarrhoea

13itching

13hyperm

enorrhoea

Polypno.4

,13

CRC,

13lung

Ca

13endometrialCa

Westet

al102

R,DB,PC

FAP

58EPA2gas

free

fattyacid

6months

No.

rectalpolyps

[EPA,DHA&AA4

[NauseaEPAgrp(31%

vs10%).

Other

SEs

422%Y

polypno,30%Y

polypsize

[,increase;Y,decrease;4

,no

change.

AA,arachidonicacid;abdo,abdominal;AI,apoptosisindex;Ca,cancer;C

,controlled;CRC,colorectalcancer;DB,doubleblind;DHA,docosahexaenoicacid;EPA

,eicosapentaenoicacid;FAP,familialadenom

atouspolyposis;FO

,fish

oil(FO

54%EPA/46%

DHA

asethylesters;FO

48%EPA/44%

DHA,as

triglycerides;FO

55%EPA/30%

DHA/15%

otheru-3

PUFA

s;FO

4¼5

8%EPA/42%

DHA;FO

20%EPA/80%

DHA;FO

21%EPA/79%

DHA);grp,

group;

mo,

month;NS,notstatistically

significant;PC

,placebo

controlled;

PG,prostaglandin;

PI,proliferationindex;

PUFA

,polyunsaturatedfattyacid;R,random

ised;SE,

side

effects;

wk,weeks.

142 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 9: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

the commonest gastrointestinal adverse event,diarrhoea, was associated with the EPA 2 g dailydose of EPA (14.0%), but not the 1 g dose (2%, cf.placebo 7.8%), suggesting dose de-escalation asa potential strategy for maintaining u-3 PUFAtreatment if diarrhoea does occur.64

The excellent safety and tolerability profile of u-3 PUFA supplementation is supported in largestudies of the effects of u-3 PUFAs in cardiovas-cular disease. In the GISSI prevention study, 2836patients were given 0.85 g u-3 PUFA for3.5 years.105 The commonest side effects weregastrointestinal disturbance (4.9%) and nausea(1.4%) Only 3.8% of patients discontinued thedrug. Gastrointestinal symptoms are typicallymild diarrhoea, indigestion, belching or a ‘fishy ’taste.106 107 In a study using a higher dose of 6.9 gEPA+DHA daily for 6 months, gastrointestinaldisturbance was reported by 7% of 275 patients,but also by 8% of patients taking placebo (corn-oil)capsules.108

For more detail, the reader is recommended toreview concise overviews of the safety and toxi-cology of EPA and DHA by Kim et al92 and Lien.109

Risk of bleedingu-3 PUFAs have antiplatelet activity, thought to bedue to a reduction in COX-1-dependent productionof thromboxane A2 from AA in platelets, leading toreduced platelet activation and aggregation.110

However, the concern that u-3 PUFAs may increasebleeding has not been realised clinically, even whenco-administered with aspirin. A recent reviewhighlighted that in over 4000 patients taking u-3PUFAs undergoing carotid endarterectomy, percu-taneous transluminal coronary angioplasty orcoronary artery bypass grafting, excess bleedingwas virtually non-existent.106 111

Environmental contaminantsWhile it has been suggested that fish oils maycontain potentially toxic levels of heavy metals (eg,mercury), environmental toxins and fat-solublevitamins, all of which can accumulate in fish, thereis no evidence to support this in practice.106 112 Theextraction and purification methods used in theproduction of fish oil supplements result in negli-gible or undetectable levels of such toxins,106 112

and so fish oil preparations given in daily doses upto 2 g/day are classified by the US Food and DrugAdministration (FDA) as ‘generally regarded assafe.’

TRANSLATIONAL STUDIES OF HUMANCOLORECTAL BIOMARKERS AND u-3 PUFACONTENTMucosal biomarker studiesThe long natural history of colorectal carcinogen-esis in humans precludes the use of CRC incidenceas a primary end point in clinical interventionstudies and a 3e5 year intervention period isgenerally required for adenoma (polyp) preventiontrials. Therefore many investigators have measured

the effect of u-3 PUFA administration on putativemucosal biomarkers of future CRC risk, such asepithelial cell mitosis frequency in microdissectedwhole crypts or immunohistochemistry for the Ki-67 ‘proliferation’ antigen. The design of suchstudies and their main findings are summarised intable 3.We identified eight studies of oral u-3 PUFA

supplementation in patients with previous‘sporadic’ colorectal adenomas (table 3), in whichcolorectal mucosal biopsy specimens were obtainedat endoscopy before and after u-3 PUFA supple-mentation. Two studies used EPA-FFA, whereas theothers used a fish oil mixture containing EPA andDHA in varying proportions in either the ethylester or triglyceride form. In six of eight studiesa 13e70% reduction in mucosal epithelial cellproliferation index was observed compared withthe respective placebo group.64 93 94 96 97 100 Inthose studies comparing different doses of u-3PUFA, a dose-dependent reduction in the PI wasobserved.94 96

By contrast, two studies demonstrated no changein PI following administration of 2.4 g u-3 PUFAdaily for 12 weeks98 or low-dose u-3 PUFA (400 mgDHA+100 mg EPA/day) for 1e2 years.99 The latterstudy did demonstrate a 50% increase in AI andincreased expression of the pro-apoptotic proteinBAX. Although in vivo preclinical work suggeststhat u-3 PUFAs may have anti-CRC activity via anincrease in apoptosis, AI has been measured in onlytwo other studies, which demonstrated a signifi-cant increase in AI after 3e6 months’ treatmentwith EPA 2 g daily.64 100 One further study noteda more modest 16% reduction in PI after 28 dayssupplementation in healthy volunteers.95 In thosestudies measuring mucosal PUFA content, signifi-cant increases in mucosal DHA and EPA, togetherwith a reduction in mucosal AA, were observed inall but one study (table 3).

Polyp prevention studiesPatients with FAP have a heterozygous germlinemutation in the Adenomatous Polyposis Coli (APC)gene. Following somatic loss of the second APCallele, multiple colorectal adenomas develop, andprophylactic colectomy is advised in order toprevent CRC. Patients who undergo total colec-tomy with ileorectal anastomosis require regularendoscopic surveillance of the remaining rectum,which remains at risk of further polyps. Studies ofpotential chemopreventative agents in thesepatients allow polyp size and number to be used asend points over a relatively short (6e12 months)period of time.A recent phase III randomised, double-blind,

placebo-controlled trial investigated treatmentwith EPA-FFA 2 g daily for 6 months in patients(n¼58) with FAP who had previously undergonecolectomy and ileorectal anastomosis.102 Rectalpolyp multiplicity and size were measured byblinded video-endoscopic assessment of a tattooedarea at baseline and at 6 months. There wasa 22.4% reduction in polyp number in the EPA

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 143

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 10: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

group compared with placebo (p¼0.01), a similarmagnitude of reduction to that seen with theselective COX-2 inhibitor celecoxib.113 In keepingwith previous studies, a significant increase inmucosal EPA levels was seen. The demonstration ofchemopreventative efficacy of EPA-FFA in patientswith FAP has led to funding of a randomised,placebo-controlled trial of EPA-FFA in patients whohave had ‘sporadic’ colorectal adenomas removedand who require further colonoscopic surveillance(http://www.eme.ac.uk/funded_projects/).Only one further polyp prevention study was

identified.101 In this small study, five patients whohad previously undergone colectomy for FAP or had>30 colorectal polyps removed endoscopically weregiven 2.2 g DHA+0.6 g EPA daily for 1e2 years.101

No significant change in polyp number was seen.One patient developed proximal CRC, a secondpatient developed lung cancer and a third patientdeveloped endometrial cancer before termination ofthe study. The published report does not make itclear whether this study was stopped prematurelyand gives no indication of the planned sample sizefor the trial.

CLINICAL TRIALS OF u-3 PUFAS IN PATIENTSWITH CRCSingle agent treatmentDespite strong in vitro and animal model evidencefor direct anti-CRC activity of u-3 PUFAs, nopublished studies have yet investigated the anti-neoplastic effect of u-3 PUFAs in patients withprimary or metastatic CRC. The authors arecurrently recruiting patients into a phase II trial ofthe safety and efficacy of oral EPA-FFA 2 g daily inpatients awaiting surgery for colorectal cancer livermetastases (CRCLM) (NCT01070355). Primarytissue biomarker end points (tumour cell prolifera-tion, apoptosis and angiogenesis) are supported bymechanistic analyses investigating whether a PGE2-PGE3 switch occurs in CRC tissue. The authors areaware of only one further trial, registered on http://clinicaltrials.org/ (NCT00942292), investigating theeffects of a parenteral fish oil preparation ontumour angiogenesis in CRCLM. u-3 fish oils arealso being investigated for the treatment ofadvanced prostatic cancer (NCT00996749), and inthe prevention and treatment of breast cancer(NCT01282580, NCT00627276).

Adjuvant therapy with traditionalchemoradiotherapyDHA has been shown to potentiate the pro-apoptotic properties of 5-fluorouracil against severalCRC cell lines, with a reduction in the expression ofthe antiapoptotic proteins BCL-2 and BCL-XL.114

Moreover, the addition of a fish oil mixture (55%EPA, 45% DHA) to 5-fluorouracil synergisticallyreduces CRC cell proliferation.115 Similar resultshave been demonstrated in rodent CRC models.116

These findings complement those of combinationfish oil treatment with other chemotherapeuticdrugs in vitro and in vivo for other solid tumourand haematological malignancies.117

We identified only one published human study ofthe anticancer effects of combining u-3 PUFAswith chemotherapy. A phase II study evaluatedaddition of 1.8 g DHA daily to an anthracycline-based chemotherapy regimen for metastatic breastcancer. Patients were dichotomised into two groupsbased on high or low DHA incorporation intoplasma phospholipids. The high DHA-incorpora-tion group had a significantly longer time to diseaseprogression (median 8.7 months vs 3.5 months)and overall survival (median 34 months vs18 months).118

There are a small number of in vitro studiesdemonstrating that both EPA and DHA potentiatethe cytotoxicity of ionising radiation in various celllines, including CRC cells.119 A retrospective reviewof 143 patients, who had been prescribed u-3 fishoil (0.9 g EPA, 1.5 g DHA daily) for 18 weeksfollowing radiotherapy for brain metastases, foundreduced radionecrosis (3.5% vs 14.1%) andimproved overall survival (median survival88.8 weeks vs 54.1 weeks) compared with the 262patients who had not been prescribed fish oil.120

While un-blinded and non-randomised, thisstudy nevertheless demonstrated a clear survivaladvantage linked to u-3 PUFA supplementation.One further study demonstrated improved

tolerability of chemoradiation for oesophagealcancer in patients taking u-3 PUFA supplements,with a reduction in the incidence of grade 2e4neutropenia, diarrhoea and pharyngitis comparedwith those not taking supplements.121 There isa clear need for further human studies to evaluatethe role of u-3 PUFA supplementation inimproving the efficacy and/or tolerability ofchemotherapy and radiotherapy. Two ongoingstudies are investigating the combination of fish oil(either enteral or parenteral) and gemcitabine inpatients with advanced pancreatic cancer (studyidentifiers NCT01019382 and JPRN-UMIN000003658). A DHA-paclitaxel compound isalso being investigated for the treatment of meta-static prostate (NCT00024414), pancreatic(NCT00024375) and CRC (NCT00024401).

u-3 PUFAs as nutritional treatmentWhile the potential anti-CRC benefits of u-3PUFAs as either single agent or combination treat-ment are yet to be established in human studies,the role of u-3 PUFA supplementation forpreventing cancer-related cachexia has been morewidely investigated.There have been two systematic reviews of the

effects of u-3 PUFAs on cancer cachexia andoutcome. A Cochrane review of the benefit of EPAfor treating cancer cachexia identified 59 potentialtrials, of which only five were randomisedcontrolled trials (RCTs).122 Patients in these studiesgenerally had pancreatic, gastrointestinal or lungcancers. No trial has studied CRC patients exclu-sively. The Cochrane review concluded that therewere insufficient data to determine whether EPAsupplementation was beneficial in the treatment ofcancer cachexia, and identified a clear need for

144 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 11: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

a large well conducted RCT. Four of the five trialsdemonstrated either stabilisation of weight loss, orweight gain, following EPA supplementation, butthis was no greater than the weight changes asso-ciated with the isocaloric control diet. However,one study did demonstrate a significant dose-dependent association between EPA supplementa-tion and weight gain when taking into consider-ation the poor tolerance of up to 18 capsules/day bysome patients.123 Of the studies reporting survivaldata, one demonstrated improved survival in theEPA-supplemented group versus placebo (median390 days vs 165 days),124 but neither of the twostudies comparing EPA supplementation with anisocaloric control intervention demonstrateda survival benefit of EPA.123 125

Colomer et al published a systematic reviewincluding the above RCTs and an additional 12prospective observational studies.126 Many of thesestudies were in patients with advanced pancreaticcancer. They concluded that there was fair evidence(grade B) to recommend u-3 PUFA supplementa-tion in patients with solid upper gastrointestinaland pancreatic tumours, with the benefits ofincreased weight, appetite and quality of life, aswell as lower postoperative morbidity.We have identified only one published (uncon-

trolled) study in patients with CRC. Treatment of23 patients with advanced CRC awaiting chemo-therapy was started with oral supplement drinkscontaining a total of 2.18 g EPA+0.92 g DHA perday for 4 weeks before and 5 weeks after irinotecan-based chemotherapy (FOLFIRI). There wasa significant improvement in weight (mean increase2.5 kg) during the period of u-3 PUFA supplemen-tation before chemotherapy, despite a fall in totalprotein and energy intake in this time, with weightgain persisting after the start of chemotherapy.127

More recently, a RCT demonstrated thatpatients undergoing oesophagectomy for cancerwho received EPA-enriched enteral nutrition for5 days pre- and 21 days postoperatively conservedfat-free mass compared with patients receivingstandard enteral nutrition. Total weight loss was1.2 kg in the EPA group versus 1.9 kg in the controlgroup (p¼0.03). The EPA group also demonstratedan attenuated stress response (measured by serumtumour necrosis factor a, interleukin 8 (interleukin10 levels) compared with controls, although therewas no difference in postoperative complica-tions.128

Animal studies have shown that u-3 PUFAsupplementation impairs the host response tomycobacterial infection.129 130 This raises thequestion whether the anti-inflammatory andimmunoregulatory effects of u-3 PUFA supple-mentation might increase susceptibility to infec-tion in immunocompromised patients. Thiscomplex topic was comprehensively examined ina review in 2002.131 Subsequent preclinical datahave been conflicting.129 130 132e135 Moreover,results from limited human observational studiesinvestigating fatty acid intake and risk of commu-nity-acquired pneumonia are inconclusive.136 137

Long-term observational studies of u-3 PUFAintake have not revealed an increase in infectiousadverse events.136 137 Further trials of u-3 PUFAsfor CRC treatment and/or prevention shouldexamine whether u-3 PUFAs alter host suscepti-bility to infection, particularly in immunocompro-mised patients with cancer who may requiresurgery.

IS THERE A DIFFERENCE IN ANTI-CRC EFFICACYBETWEEN EPA AND DHA?It is clear from the preclinical ‘prevention’ studiesthat combination EPA and DHA treatment hasanti-CRC activity. There is only one ‘head-to-head’comparison, which demonstrated that EPA hadgreater efficacy than DHA.60 However, the twopreclinical ‘treatment’ studies that have beenpublished did not show any difference between EPAand DHA.71 79 As far as we are aware, there is nopublished report comparing EPA and DHA inpatients with, or at-risk for, CRC. It is also unclearwhether an EPA/DHA mixture would give addi-tional benefits over and above equivalent doses ofeither u-3 PUFA alone.In addition, a note of caution should be made of

the two studies that demonstrated negative effectsof high-dose fish oil in a model of CRCLM (12%EPA/8% DHA mix)87 and a model of infection-induced, colitis-associated cancer (6% DHA),57 inconflict with the majority of u-3 PUFA studies. Themechanistic basis of a possible negative effect ofhigh-dose u-3 PUFAs is not clear and warrantsfurther investigation with particular emphasis onthe safe maximal dose of either EPA or DHA assingle agents, or in combination with otherchemotherapeutic agents.Any comparison between the efficacy of EPA and

DHA may also be confounded by interconversionbetween these two u-3 PUFAs in vivo. EPA to DHAconversion can occur, via docosapentaenoic acid, ina two-stage process involving elongase and desa-turase activity (with docosapentaenoic acideEPAconversion being the limiting step). However,limited evidence from clinical trials suggests thatthere is no significant EPAeDHA conversion inhuman colorectal mucosa.100 102 By contrast,administration of DHA in humans has been shownto result in an increase in EPA content in plasmaphospholipids.138 This may be as high as 12%during chronic dosing with DHA and is thought tooccur via b-oxidation of the fatty acid.138 Thepossibility that DHA treatment could result in aneventual increase in EPA content of normal and/orneoplastic colorectal mucosa requires investigation.

CONCLUSIONu-3 PUFAs are likely to have multifaceted roles inboth prevention and treatment of CRC. Theexcellent tolerability and safety profile of u-3PUFAs combined with other health benefits,particularly cardiovascular, make u-3 PUFAs anattractive candidate for prevention and treatmentof CRC (and other cancers).

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 145

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 12: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

There is a wealth of evidence to support theantineoplastic effects of u-3 PUFAs, with both invivo models and human biomarker studiesdemonstrating a clear reduction in mucosal prolif-eration and an increase in mucosal apoptosis, aswell as mucosal u-3 PUFA incorporation at theexpense of u-6 PUFA. The role of u-3 PUFAs inprevention of CRC is further supported by therecent demonstration of chemopreventative effi-cacy of EPA-FFA in patients with FAP.102

Strong in vivo evidence also supports a role for u-3 PUFAs in the treatment of CRC, with studiesconsistently demonstrating tumour u-3 PUFAincorporation and a reduction in tumour size. PhaseII and phase III evaluations of u-3 PUFAs arejustified to establish their safety, tolerability andantineoplastic activity in patients with establishedCRC. Ongoing studies are investigating the effectof u-3 PUFAs on tumour growth and vascularity inCRCLM. These studies should eventually informthe design of large, multicentre phase III evaluationof u-3 PUFAs in patients with CRC, both asneoadjuvant/adjuvant treatment in patientsundergoing surgery with curative intent and in themanagement of incurable disease. u-3 PUFA treat-ment should be investigated, as a single agent andin combination with other chemotherapeuticagents, for treatment of CRC. Evaluation ofcombination therapy with u-3 PUFAs is alreadyunderway in other cancers, including breast andpancreatic cancer.A third potential role of u-3 PUFAs as nutritional

treatment in patients with CRC has also beenidentified, with reported improvements in cancer-related cachexia and perioperative outcomes inother cancers.Improved understanding of the mechanisms of

anti-CRC activity of EPA and DHA will lead toidentification and validation of predictive thera-peutic response biomarkers for use in future clinicaltrials and help to determine the best anticanceragents to use in combination with u-3 PUFAs forprevention and/or treatment of CRC.

Competing interests MAH has received an unrestricted scientificgrant from, and performed paid consultancy work for, SLA PharmaAG, which produces a formulation of eicosapentaenoic acid.

Provenance and peer review Commissioned; externally peerreviewed.

REFERENCES1. Burdge G. Alpha-linolenic acid metabolism in men and women:

nutritional and biological implications. Curr Opin Clin Nutr MetabCare 2004;7:137e44.

2. Smith WL. Cyclooxygenases, peroxide tone and the allure of fishoil. Curr Opin Cell Biol 2005;17:174e82.

3. Hyde CA, Missailidis S. Inhibition of arachidonic acid metabolismand its implication on cell proliferation and tumour-angiogenesis.Int Immunopharmacol 2009;9:701e15.

4. Chapkin RS, McMurray DN, Lupton JR. Colon cancer, fatty acidsand anti-inflammatory compounds. Curr Opin Gastroenterol2007;23:48e54.

5. Roth EM, Harris WS. Fish oil for primary and secondaryprevention of coronary heart disease. Curr Atheroscler Rep2010;12:66e72.

6. Saravanan P, Davidson NC, Schmidt EB, et al. Cardiovasculareffects of marine omega-3 fatty acids. Lancet 2010;376:540e50.

7. Fetterman JW Jr, Zdanowicz MM. Therapeutic potential of n-3polyunsaturated fatty acids in disease. Am J Health Syst Pharm2009;66:1169e79.

8. Calviello G, Serini S, Piccioni E. n-3 polyunsaturated fatty acidsand the prevention of colorectal cancer: molecular mechanismsinvolved. Curr Med Chem 2007;14:3059e69.

9. Hull MA, Ko SC, Hawcroft G. Prostaglandin EP receptors: targetsfor treatment and prevention of colorectal cancer? Mol CancerTher 2004;3:1031e9.

10. Hawcroft G, Loadman PM, Belluzzi A, et al. Effect ofeicosapentaenoic acid on E-type prostaglandin synthesis and EP4receptor signalling in human colorectal cancer cells. Neoplasia2010;12:618e27.

11. Yang P, Chan D, Felix E, et al. Formation and antiproliferativeeffect of prostaglandin E(3) from eicosapentaenoic acid in humanlung cancer cells. J Lipid Res 2004;45:1030e9.

12. Vanamala J, Glagolenko A, Yang P, et al. Dietary fish oil andpectin enhance colonocyte apoptosis in part through suppressionof PPAR delta/PGE(2) and elevation of PGE(3). Carcinogenesis2008;29:790e6.

13. Vecchio AJ, Simmons DM, Malkowski MG. Structural basis offatty acid substrate binding to cyclooxygenase-2. J Biol Chem2010;285:22152e63.

14. Yaqoob P. The nutritional significance of lipid rafts. Annu RevNutr 2009;29:257e82.

15. Schley PD, Brindley DN, Field CJ. (n-3) PUFA alter raft lipidcomposition and decrease epidermal growth factor receptorlevels in lipid rafts of human breast cancer cells. J Nutr2007;137:548e53.

16. Chapkin RS, Hong MY, Fan YY, et al. Dietary n-3 PUFA altercolonocyte mitochondrial membrane composition and function.Lipids 2002;37:193e9.

17. Collett ED, Davidson LA, Fan YY, et al. n-6 and n-3polyunsaturated fatty acids differentially modulate oncogenic Rasactivation in colonocytes. Am J Physiol Cell Physiol 2001;280:C1066e75.

18. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygenspecies (ROS) in apoptosis induction. Apoptosis 2000;5:415e18.

19. Chang WL, Chapkin RS, Lupton JR. Fish oil blocksazoxymethane-induced rat colon tumorigenesis by increasing celldifferentiation and apoptosis rather than decreasing cellproliferation. J Nutr 1998;128:491e7.

20. Crim KC, Sanders LM, Hong MY, et al. Upregulation of p21Waf1/Cip1 expression in vivo by butyrate administration can bechemoprotective or chemopromotive depending on the lipidcomponent of the diet. Carcinogenesis 2008;29:1415e20.

21. Sanders LM, Henderson CE, Hong MY, et al. An increase inreactive oxygen species by dietary fish oil coupled with theattenuation of antioxidant defenses by dietary pectin enhancesrat colonocyte apoptosis. J Nutr 2004;134:3233e8.

22. Kolar SS, Barhoumi R, Callaway ES, et al. Synergy betweendocosahexaenoic acid and butyrate elicits p53-independentapoptosis via mitochondrial Ca(2+) accumulation in colonocytes.Am J Physiol Gastrointest Liver Physiol 2007;293:G935e43.

23. Kolar SS, Barhoumi R, Lupton JR, et al. Docosahexaenoic acidand butyrate synergistically induce colonocyte apoptosis byenhancing mitochondrial Ca2+ accumulation. Cancer Res2007;67:5561e8.

24. Ng Y, Barhoumi R, Tjalkens RB, et al. The role ofdocosahexaenoic acid in mediating mitochondrial membrane lipidoxidation and apoptosis in colonocytes. Carcinogenesis2005;26:1914e21.

25. Buchanan FG, Wang D, Bargiacchi F, et al. Prostaglandin E2regulates cell migration via the intracellular activation of theepidermal growth factor receptor. J Biol Chem2003;278:35451e7.

26. Jump DB. Fatty acid regulation of gene transcription. Crit RevClin Lab Sci 2004;41:41e78.

27. Serhan CN, Arita M, Hong S, et al. Resolvins, docosatrienes,and neuroprotectins, novel omega-3-derived mediators, and theirendogenous aspirin-triggered epimers. Lipids 2004;39:1125e32.

28. Arita M, Bianchini F, Aliberti J, et al. Stereochemicalassignment, antiinflammatory properties, and receptor for theomega-3 lipid mediator resolvin E1. J Exp Med.2005;201:713e22.

29. Oh SF, Pillai PS, Recchiuti A, et al. Pro-resolving actions andstereoselective biosynthesis of 18S E-series resolvins in humanleukocytes and murine inflammation. J Clin Invest2011;121:569e81.

30. Arita M, Clish CB, Serhan CN. The contributions of aspirin andmicrobial oxygenase to the biosynthesis of anti-inflammatoryresolvins: novel oxygenase products from omega-3

146 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 13: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

polyunsaturated fatty acids. Biochem Biophys Res Commun2005;338:149e57.

31. Hong S, Gronert K, Devchand PR, et al. Novel docosatrienes and17S-resolvins generated from docosahexaenoic acid in murinebrain, human blood, and glial cells. Autacoids in anti-inflammation. J Biol Chem 2003;278:14677e87.

32. Serhan CN, Yang R, Martinod K, et al. Maresins: novelmacrophage mediators with potent antiinflammatory andproresolving actions. J Exp Med 2009;206:15e23.

33. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation:dual anti-inflammatory and pro-resolution lipid mediators. Nat RevImmunol 2008;8:349e61.

34. Campbell EL, MacManus CF, Kominsky DJ, et al. ResolvinE1-induced intestinal alkaline phosphatase promotes resolution ofinflammation through LPS detoxification. Proc Natl Acad Sci U S A2010;107:14298e303.

35. Clevers H. At the crossroads of inflammation and cancer. Cell2004;118:671e4.

36. Calder PC. Immunomodulation by omega-3 fatty acids.Prostaglandins Leukot Essent Fatty Acids 2007;77:327e35.

37. Szymczak M, Murray M, Petrovic N. Modulation of angiogenesisby omega-3 polyunsaturated fatty acids is mediated bycyclooxygenases. Blood 2008;111:3514e21.

38. Tsuzuki T, Shibata A, Kawakami Y, et al. Conjugatedeicosapentaenoic acid inhibits vascular endothelial growth factor-induced angiogenesis by suppressing the migration of humanumbilical vein endothelial cells. J Nutr 2007;137:641e6.

39. Byers T. Nutrition and cancer among American Indians andAlaska Natives. Cancer 1996;78:1612e16.

40. World Cancer Research Fund/American Institute forCancer Research. Food, Nutrition, Physical Activity, and thePrevention of Cancer: a Global Perspective. American Institute ofCancer Research (AICR), Washington, DC: 2007.

41. Geelen A, Schouten JM, Kamphuis C, et al. Fish consumption,n-3 fatty acids, and colorectal cancer: a meta-analysis ofprospective cohort studies. Am J Epidemiol 2007;166:1116e25.

42. Kim S, Sandler DP, Galanko J, et al. Intake of polyunsaturatedfatty acids and distal large bowel cancer risk in whites andAfrican Americans. Am J Epidemiol 2010;171:969e79.

43. Pot GK, Geelen A, van Heijningen EM, et al. Opposingassociations of serum n-3 and n-6 polyunsaturated fatty acidswith colorectal adenoma risk: an endoscopy-based case-controlstudy. Int J Cancer 2008;123:1974e7.

44. Nelson RL, Tanure JC, Andrianopoulos G, et al. A comparison ofdietary fish oil and corn oil in experimental colorectalcarcinogenesis. Nutr Cancer 1988;11:215e20.

45. Minoura T, Takata T, Sakaguchi M, et al. Effect of dietaryeicosapentaenoic acid on azoxymethane-induced coloncarcinogenesis in rats. Cancer Res 1988;48:4790e4.

46. Reddy BS, Sugie S. Effect of different levels of omega-3 andomega-6 fatty acids on azoxymethane-induced coloncarcinogenesis in F344 rats. Cancer Res 1988;48:6642e7.

47. Deschner EE, Lytle JS, Wong G, et al. The effect of dietaryomega-3 fatty acids (fish oil) on azoxymethanol-induced focal areasof dysplasia and colon tumor incidence. Cancer 1990;66:2350e6.

48. Reddy BS, Burill C, Rigotty J. Effect of diets high in omega-3 andomega-6 fatty acids on initiation and postinitiation stages of coloncarcinogenesis. Cancer Res 1991;51:487e91.

49. Takahashi M, Minamoto T, Yamashita N, et al. Reduction information and growth of 1,2-dimethylhydrazine-induced aberrantcrypt foci in rat colon by docosahexaenoic acid. Cancer Res1993;53:2786e9.

50. Hendrickse CW, Keighley MR, Neoptolemos JP. Dietaryomega-3 fats reduce proliferation and tumor yields at colorectalanastomosis in rats. Gastroenterology 1995;109:431e9.

51. Takahashi M, Fukutake M, Isoi T, et al. Suppression ofazoxymethane-induced rat colon carcinoma development bya fish oil component, docosahexaenoic acid (DHA).Carcinogenesis 1997;18:1337e42.

52. Good CK, Lasko CM, Adam J, et al. Diverse effect of fish oil onthe growth of aberrant crypt foci and tumor multiplicity in F344rats. Nutr Cancer 1998;31:204e11.

53. Singh J, Hamid R, Reddy BS. Dietary fish oil inhibits theexpression of farnesyl protein transferase and colon tumordevelopment in rodents. Carcinogenesis 1998;19:985e9.

54. Latham P, Lund EK, Johnson IT. Dietary n-3 PUFA increases theapoptotic response to 1,2-dimethylhydrazine, reduces mitosis andsuppresses the induction of carcinogenesis in the rat colon.Carcinogenesis 1999;20:645e50.

55. Rao CV, Hirose Y, Indranie C, et al. Modulation of experimentalcolon tumorigenesis by types and amounts of dietary fatty acids.Cancer Res 2001;61:1927e33.

56. Moreira AP, Sabarense CM, Dias CM, et al. Fish oil ingestionreduces the number of aberrant crypt foci and adenoma in1,2-dimethylhydrazine-induced colon cancer in rats. Braz J MedBiol Res 2009;42:1167e72.

57. Woodworth HL, McCaskey SJ, Duriancik DM, et al. Dietary fishoil alters T lymphocyte cell populations and exacerbates diseasein a mouse model of inflammatory colitis. Cancer Res2010;70:7960e9.

58. Oshima M, Takahashi M, Oshima H, et al. Effects ofdocosahexaenoic acid (DHA) on intestinal polyp development inApc delta 716 knockout mice. Carcinogenesis 1995;16:2605e7.

59. Paulsen JE, Elvsaas IK, Steffensen IL, et al. A fish oil derivedconcentrate enriched in eicosapentaenoic and docosahexaenoicacid as ethyl ester suppresses the formation and growth ofintestinal polyps in the Min mouse. Carcinogenesis1997;18:1905e10.

60. Petrik MBH, McEntee MF, Johnson BT, et al. Highly unsaturated(n-3) fatty acids, but not alpha-linolenic, conjugated linoleic orgamma-linolenic acids, reduce tumorigenesis in Apc(Min/+)mice. J Nutr 2000;130:2434e43.

61. Petrik MBH, McEntee MF, Chiu CH, et al. Antagonism ofarachidonic acid is linked to the antitumorigenic effect of dietaryeicosapentaenoic acid in Apc(Min/+) mice. J Nutr2000;130:1153e8.

62. Bose M, Hao X, Ju J, et al. Inhibition of tumorigenesis inApcMin/+ mice by a combination of (-)-epigallocatechin-3-gallate and fish oil. J Agric Food Chem 2007;55:7695e700.

63. Fini L, Piazzi G, Ceccarelli C, et al. Highly purifiedeicosapentaenoic acid as free fatty acids strongly suppressespolyps in Apc (Min/+) mice. Clin Cancer Res 2010;16:5703e11.

64. West NJ, Belluzzi A, Lund EK, et al. Eicosapentaenoic acid(EPA), as the free fatty acid, reduces colonic crypt cellproliferation in patients with sporadic colorectal polyps. Gut2009;58:A68.

65. Mund RC, Pizato N, Bonatto S, et al. Decreased tumor growth inWalker 256 tumor-bearing rats chronically supplemented withfish oil involves COX-2 and PGE2 reduction associated withapoptosis and increased peroxidation. Prostaglandins LeukotEssent Fatty Acids 2007;76:113e20.

66. Davidson LA, Nguyen DV, Hokanson RM, et al.Chemopreventive n-3 polyunsaturated fatty acids reprogramgenetic signatures during colon cancer initiation and progressionin the rat. Cancer Res 2004;64:6797e804.

67. Boudreau MD, Sohn KH, Rhee SH, et al. Suppression of tumorcell growth both in nude mice and in culture by n-3polyunsaturated fatty acids: mediation through cyclooxygenase-independent pathways. Cancer Res 2001;61:1386e91.

68. Clarke RG, Lund EK, Latham P, et al. Effect of eicosapentaenoicacid on the proliferation and incidence of apoptosis in thecolorectal cell line HT29. Lipids 1999;34:1287e95.

69. Llor X, Pons E, Roca A, et al. The effects of fish oil, olive oil, oleicacid and linoleic acid on colorectal neoplastic processes. Clin Nutr2003;22:71e9.

70. Tsai WS, Nagawa H, Kaizaki S, et al. Inhibitory effects of n-3polyunsaturated fatty acids on sigmoid colon cancertransformants. J Gastroenterol 1998;33:206e12.

71. Calviello G, Di Nicuolo F, Gragnoli S, et al. n-3 PUFAs reduceVEGF expression in human colon cancer cells modulating theCOX-2/PGE2 induced ERK-1 and -2 and HIF-1alpha inductionpathway. Carcinogenesis 2004;25:2303e10.

72. Chen ZY, Istfan NW. Docosahexaenoic acid is a potent inducerof apoptosis in HT-29 colon cancer cells. Prostaglandins LeukotEssent Fatty Acids 2000;63:301e8.

73. Mengeaud V, Nano JL, Fournel S, et al. Effects ofeicosapentaenoic acid, gamma-linolenic acid and prostaglandinE1 on three human colon carcinoma cell lines. ProstaglandinsLeukot Essent Fatty Acids 1992;47:313e19.

74. Palozza P, Calviello G, Maggiano N, et al. Beta-caroteneantagonizes the effects of eicosapentaenoic acid on cell growthand lipid peroxidation in WiDr adenocarcinoma cells. Free RadicBiol Med 2000;28:228e34.

75. Calviello G, Resci F, Serini S, et al. Docosahexaenoic acidinduces proteasome-dependent degradation of beta-catenin,down-regulation of survivin and apoptosis in human colorectalcancer cells not expressing COX-2. Carcinogenesis2007;28:1202e9.

76. Lee SA, Kim HJ, Chang KC, et al. DHA and EPA Down-regulateCOX-2 Expression through Suppression of NF-kappaB Activity inLPS-treated Human Umbilical Vein Endothelial Cells. Korean JPhysiol Pharmacol 2009;13:301e7.

77. Paulsen JE, Stamm T, Alexander J. A fish oil-derivedconcentrate enriched in eicosapentaenoic and docosahexaenoic

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 147

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 14: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

acid as ethyl esters inhibits the formation and growth of aberrantcrypt foci in rat colon. Pharmacol Toxicol 1998;82:28e33.

78. Cannizzo F Jr, Broitman SA. Postpromotional effects of dietarymarine or safflower oils on large bowel or pulmonary implants ofCT-26 in mice. Cancer Res 1989;49:4289e94.

79. Iigo M, Nakagawa T, Ishikawa C, et al. Inhibitory effects ofdocosahexaenoic acid on colon carcinoma 26 metastasis to thelung. Br J Cancer 1997;75:650e5.

80. Calder PC, Davis J, Yaqoob P, et al. Dietary fish oil suppresseshuman colon tumour growth in athymic mice. Clin Sci1998;94:303e11.

81. Kato T, Hancock RL, Mohammadpour H, et al. Influence ofomega-3 fatty acids on the growth of human colon carcinoma innude mice. Cancer Lett 2002;187:169e77.

82. Togni V, Ota CCC, Folador A, et al. Cancer cachexia and tumorgrowth reduction in Walker 256 tumor-bearing rats supplementedwith N-3 polyunsaturated fatty acids for one generation. NutrCancer 2003;46:52e8.

83. Tsuzuki T, Igarashi M, Miyazawa T. Conjugatedeicosapentaenoic acid (EPA) inhibits transplanted tumor growthvia membrane lipid peroxidation in nude mice. J Nutr2004;134:1162e6.

84. Pizato N, Bonatto S, Yamazaki RK, et al. Ratio of n6 to n-3 fattyacids in the diet affects tumor growth and cachexia in Walker256 tumor-bearing rats. Nutr Cancer 2005;53:194e201.

85. Iwamoto S, Senzaki H, Kiyozuka Y, et al. Effects of fatty acids onliver metastasis of ACL-15 rat colon cancer cells. Nutr Cancer1998;31:143e50.

86. Gutt CN, Brinkmann L, Mehrabi A, et al. Dietary omega-3-polyunsaturated fatty acids prevent the development ofmetastases of colon carcinoma in rat liver. Eur J Nutr2007;46:279e85.

87. Griffini P, Fehres O, Klieverik L, et al. Dietary omega-3polyunsaturated fatty acids promote colon carcinoma metastasisin rat liver. Cancer Res 1998;58:3312e19.

88. Beckermann B, Beneke M, Seitz I. Comparative bioavailability ofeicosapentaenoic acid and docasahexaenoic acid fromtriglycerides, free fatty acids and ethyl esters in volunteers.Arzneimittelforschung 1990;40:700e4.

89. Lawson LD, Hughes BG. Human absorption of fish oil fatty acidsas triacylglycerols, free acids, or ethyl esters. Biochem BiophysRes Commun 1988;152:328e35.

90. Yang LY, Kuksis A, Myher JJ. Lipolysis of menhaden oiltriacylglycerols and the corresponding fatty acid alkyl esters bypancreatic lipase in vitro: a reexamination. J Lipid Res1990;31:137e47.

91. Ackman RG. The absorption of fish oils and concentrates. Lipids1992;27:858e62.

92. Kim W, McMurray DN, Chapkin RS. n-3 polyunsaturated fattyacidsephysiological relevance of dose. Prostaglandins LeukotEssent Fatty Acids 2010;82:155e8.

93. Anti M, Marra G, Armelao F, et al. Effect of omega-3 fatty acidson rectal mucosal cell proliferation in subjects at risk for coloncancer. Gastroenterology 1992;103:883e91.

94. Bartoli GM, Palozza P, Marra G, et al. n-3 PUFA and alpha-tocopherol control of tumor cell proliferation. Mol Aspects Med1993;14:247e52.

95. Bartram HP, Gostner A, Scheppach W, et al. Effects of fish oilon rectal cell proliferation, mucosal fatty acids, and prostaglandinE2 release in healthy subjects. Gastroenterology1993;105:1317e22.

96. Anti M, Armelao F, Marra G, et al. Effects of different doses offish oil on rectal cell proliferation in patients with sporadic colonicadenomas. Gastroenterology 1994;107:1709e18.

97. Huang YC, Jessup JM, Forse RA, et al. n-3 fatty acids decreasecolonic epithelial cell proliferation in high-risk bowel mucosa.Lipids 1996;31:S313e17.

98. Gee JM, Watson M, Matthew JA, et al. Consumption of fish oilleads to prompt incorporation of eicosapentaenoic acid intocolonic mucosa of patients prior to surgery for colorectal cancer,but has no detectable effect on epithelial cytokinetics. J Nutr1999;129:1862e5.

99. Cheng J, Ogawa K, Kuriki K, et al. Increased intake of n-3polyunsaturated fatty acids elevates the level of apoptosis in thenormal sigmoid colon of patients polypectomized for adenomas/tumors. Cancer Lett 2003;193:17e24.

100. Courtney ED, Matthews S, Finlayson C, et al. Eicosapentaenoicacid (EPA) reduces crypt cell proliferation and increasesapoptosis in normal colonic mucosa in subjects with a history ofcolorectal adenomas. Int J Colorectal Dis 2007;22:765e76.

101. Akedo I, Ishikawa H, Nakamura T, et al. Three cases withfamilial adenomatous polyposis diagnosed as having malignant

lesions in the course of a long-term trial using docosahexanoicacid (DHA)-concentrated fish oil capsules. Jpn J Clin Oncol1998;28:762e5.

102. West NJ, Clark SK, Phillips RK, et al. Eicosapentaenoic acidreduces rectal polyp number and size in familial adenomatouspolyposis. Gut 2010;59:918e25.

103. Gibney MJ, Hunter B. The effects of short- and long-termsupplementation with fish oil on the incorporation of n-3polyunsaturated fatty acids into cells of the immune system inhealthy volunteers. Eur J Clin Nutr 1993;47:255e9.

104. Hillier K, Jewell R, Dorrell L, et al. Incorporation of fatty acidsfrom fish oil and olive oil into colonic mucosal lipids and effectsupon eicosanoid synthesis in inflammatory bowel disease. Gut1991;32:1151e5.

105. GISSI. Dietary supplementation with n-3 polyunsaturated fattyacids and vitamin E after myocardial infarction: results of theGISSI-Prevenzione trial. Gruppo Italiano per lo Studio dellaSopravvivenza nell’Infarto miocardico. Lancet 1999;354:447e55.

106. Bays H. Clinical overview of Omacor: a concentrated formulationof omega-3 polyunsaturated fatty acids. Am J Cardiol2006;98:71ie6i.

107. Kris-Etherton PM, Harris WS, Appel LJ. Fish consumption, fishoil, omega-3 fatty acids, and cardiovascular disease. Circulation2002;106:2747e57.

108. Leaf A, Jorgensen MB, Jacobs AK, et al. Do fish oils preventrestenosis after coronary angioplasty? Circulation1994;90:2248e57.

109. Lien EL. Toxicology and safety of DHA. Prostaglandins LeukotEssent Fatty Acids 2009;81:125e32.

110. Dyerberg J, Bang HO, Stoffersen E, et al. Eicosapentaenoic acidand prevention of thrombosis and atherosclerosis? Lancet1978;2:117e19.

111. Harris WS. Expert opinion: omega-3 fatty acids and bleeding-cause for concern? Am J Cardiol 2007;99:44Ce6C.

112. Foran SE, Flood JG, Lewandrowski KB. Measurement ofmercury levels in concentrated over-the-counter fish oilpreparations: is fish oil healthier than fish? Arch Pathol Lab Med2003;127:1603e5.

113. Steinbach G, Lynch PM, Phillips RK, et al. The effect ofcelecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatouspolyposis. N Engl J Med 2000;342:1946e52.

114. Calviello G, Di Nicuolo F, Serini S, et al. Docosahexaenoic acidenhances the susceptibility of human colorectal cancer cells to 5-fluorouracil. Cancer Chemother Pharmacol 2005;55:12e20.

115. Jordan A, Stein J. Effect of an omega-3 fatty acid containinglipid emulsion alone and in combination with 5-fluorouracil (5-FU)on growth of the colon cancer cell line Caco-2. Eur J Nutr2003;42:324e31.

116. Wynter MP, Russell ST, Tisdale MJ. Effect of n-3 fatty acids onthe antitumour effects of cytotoxic drugs. In Vivo2004;18:543e7.

117. Calviello G, Serini S, Piccioni E, et al. Antineoplastic effects ofn-3 polyunsaturated fatty acids in combination with drugs andradiotherapy: preventive and therapeutic strategies. Nutr Cancer2009;61:287e301.

118. Bougnoux P, Hajjaji N, Ferrasson MN, et al. Improving outcomeof chemotherapy of metastatic breast cancer bydocosahexaenoic acid: a phase II trial. Br J Cancer2009;101:1978e85.

119. Benais-Pont G, Dupertuis YM, Kossovsky MP, et al. Omega-3polyunsaturated fatty acids and ionizing radiation: combinedcytotoxicity on human colorectal adenocarcinoma cells. Nutrition2006;22:931e9.

120. Gramaglia A, Loi GF, Mongioj V, et al. Increased survival in brainmetastatic patients treated with stereotactic radiotherapy,omega three fatty acids and bioflavonoids. Anticancer Res1999;19:5583e6.

121. Minami Y, Miyata H, Doki Y, et al. omega-3 Fatty acid-containingdiet (Racol) reduces toxicity of chemoradiation therapy forpatients with esophageal cancer. Gan To Kagaku Ryoho2008;35:437e40.

122. Dewey A, Baughan C, Dean T, et al. Eicosapentaenoic acid(EPA, an omega-3 fatty acid from fish oils) for the treatment ofcancer cachexia. Cochrane Database Syst Rev 2007;(4):CD004597.

123. Fearon KC, Von Meyenfeldt MF, Moses AG, et al. Effect ofa protein and energy dense N-3 fatty acid enriched oralsupplement on loss of weight and lean tissue in cancer cachexia:a randomised double blind trial. Gut 2003;52:1479e86.

124. Gogos CA, Ginopoulos P, Salsa B, et al. Dietary omega-3polyunsaturated fatty acids plus vitamin E restoreimmunodeficiency and prolong survival for severely ill patients

148 Gut 2012;61:135e149. doi:10.1136/gut.2010.233718

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from

Page 15: Recent advances in basic science Omega-3 polyunsaturated ... › content › gutjnl › 61 › 1 › 135.full.pdfdirectly or via changes in COX metabolism, thereby altering levels

with generalized malignancy: a randomized control trial. Cancer1998;82:395e402.

125. Jatoi A, Rowland K, Loprinzi CL, et al. An eicosapentaenoic acidsupplement versus megestrol acetate versus both for patientswith cancer-associated wasting: a North Central CancerTreatment Group and National Cancer Institute of Canadacollaborative effort. J Clin Oncol 2004;22:2469e76.

126. Colomer R, Moreno-Nogueira JM, Garcia-Luna PP, et al. N-3fatty acids, cancer and cachexia: a systematic review of theliterature. Br J Nutr 2007;97:823e31.

127. Read JA, Beale PJ, Volker DH, et al. Nutrition intervention usingan eicosapentaenoic acid (EPA)-containing supplement inpatients with advanced colorectal cancer. Effects on nutritionaland inflammatory status: a phase II trial. Support Care Cancer2007;15:301e7.

128. Ryan AM, Reynolds JV, Healy L, et al. Enteral nutrition enrichedwith eicosapentaenoic acid (EPA) preserves lean body massfollowing esophageal cancer surgery: results of a double-blindedrandomized controlled trial. Ann Surg 2009;249:355e63.

129. Bonilla DL, Fan YY, Chapkin RS, et al. Transgenic mice enrichedin omega-3 fatty acids are more susceptible to pulmonarytuberculosis: impaired resistance to tuberculosis in fat-1 mice.J Infect Dis 2010;201:399e408.

130. McFarland CT, Fan YY, Chapkin RS, et al. Dietary polyunsaturatedfatty acids modulate resistance to Mycobacterium tuberculosis inguinea pigs. J Nutr 2008;138:2123e8.

131. Anderson M, Fritsche KL. (n-3) Fatty acids and infectiousdisease resistance. J Nutr 2002;132:3566e76.

132. Jordao L, Lengeling A, Bordat Y, et al. Effects of omega-3 and-6 fatty acids on Mycobacterium tuberculosis in macrophagesand in mice. Microbes Infect 2008;10:1379e86.

133. Pierre M, Husson MO, Le Berre R, et al. Omega-3polyunsaturated fatty acids improve host response in chronicPseudomonas aeruginosa lung infection in mice. Am J PhysiolLung Cell Mol Physiol 2007;292:L1422e31.

134. Saini A, Harjai K, Chhibber S. Sea-cod oil supplementationalters the course of Streptococcus pneumoniae infectionin BALB/c mice. Eur J Clin Microbiol Infect Dis2011;30:393e400.

135. Tiesset H, Pierre M, Desseyn JL, et al. Dietary (n-3)polyunsaturated fatty acids affect the kinetics of pro- andantiinflammatory responses in mice with Pseudomonasaeruginosa lung infection. J Nutr 2009;139:82e9.

136. Alperovich M, Neuman MI, Willett WC, et al. Fatty acid intakeand the risk of community-acquired pneumonia in U.S. women.Nutrition 2007;23:196e202.

137. Merchant AT, Curhan GC, Rimm EB, et al. Intake of n-6 and n-3fatty acids and fish and risk of community-acquired pneumonia inUS men. Am J Clin Nutr 2005;82:668e74.

138. Arterburn LM, Hall EB, Oken H. Distribution, interconversion,and dose response of n-3 fatty acids in humans. Am J Clin Nutr2006;83:1467Se76S.

clinicians • medical students • nurses • healthcare practitioners

Have confi dence in your decision making.

The best clinical decision support tool is now available as an app for your iPhone.Visit bestpractice.bmj.com/app

FROM THE BMJ EVIDENCE CENTRE

RECEIVE

20FREE SAMPLETOPICS WHEN YOU PURCHASE

Gut 2012;61:135e149. doi:10.1136/gut.2010.233718 149

Recent advances in basic science

on July 14, 2020 by guest. Protected by copyright.

http://gut.bmj.com

/G

ut: first published as 10.1136/gut.2010.233718 on 13 April 2011. D

ownloaded from


Recommended