suppression of skeletal muscle turnover in cancer cachexia ... · begins with the association of...

12
Human Cancer Biology Suppression of Skeletal Muscle Turnover in Cancer Cachexia: Evidence from the Transcriptome in Sequential Human Muscle Biopsies Iain J. Gallagher 1 , Nathan A. Stephens 1 , Alisdair J. MacDonald 1 , Richard J.E. Skipworth 1 , Holger Husi 1 , Carolyn A. Greig 1 , James A. Ross 1 , James A. Timmons 2 , and Kenneth C.H. Fearon 1 Abstract Purpose: The mechanisms underlying muscle wasting in patients with cancer remain poorly understood, and consequently there remains an unmet clinical need for new biomarkers and treatment strategies. Experimental Design: Microarrays were used to examine the transcriptome in single biopsies from healthy controls (n ¼ 6) and in paired biopsies [pre-resection baseline (weight-loss 7%) and 8 month post- resection follow-up (disease-free/weight-stable for previous 2 months)] from quadriceps muscle of patients with upper gastrointestinal cancer (UGIC; n ¼ 12). Results: Before surgery, 1,868 genes were regulated compared with follow-up (false discovery rate, 6%). Ontology analysis showed that regulated genes belonged to both anabolic and catabolic biologic processes with overwhelming downregulation in baseline samples. No literature-derived genes from preclinical cancer cachexia models showed higher expression in baseline muscle. Comparison with healthy control muscle (n ¼ 6) revealed that despite differences in the transcriptome at baseline (941 genes regulated), the muscle of patients at follow-up was similar to control muscle (2 genes regulated). Physical activity (step count per day) did not differ between the baseline and follow-up periods (P ¼ 0.9), indicating that gene expression differences reflected the removal of the cancer rather than altered physical activity levels. Comparative gene expression analysis using exercise training signatures supported this interpretation. Conclusions: Metabolic and protein turnover–related pathways are suppressed in weight-losing patients with UGIC whereas removal of the cancer appears to facilitate a return to a healthy state, independent of changes in the level of physical activity. Clin Cancer Res; 18(10); 2817–27. Ó2012 AACR. Introduction Cancer cachexia has been defined as "a multifactorial syndrome characterized by an ongoing loss of skeletal muscle mass that cannot be fully reversed by conventional nutritional support and that leads to progressive functional impairment" (1). Cachexia impacts negatively on the qual- ity of life of patients with cancer, response to treatment and survival, and thus tackling cachexia should be a central component of patient treatment. Skeletal muscle mass is maintained by a balance between protein synthesis and degradation, and these, in turn, are principally regulated by physiologic inputs such as nutritional status and physical activity. Several systems contribute to intracellular protein breakdown including the ubiquitin/proteasome pathway (UPP), autophagy (lysosomal pathway), caspases, cathe- psins, and calcium-dependent calpains. Protein synthesis begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor 2 (eIF2), which, in turn, is influenced by external signals potentially integrated through the mTOR (2). The balance of activity of these processes represents the molec- ular basis for muscle mass homeostasis. Preclinical models of muscle atrophy, which tend to reflect acute and rapid changes in muscle mass, have highlighted the role of the UPP with increased mRNA expression of 2 muscle-specific E3 ubiquitin ligases, MuRF-1/Trim63, and MAFbx/atrogin-1/Fbxo32 (3, 4) and evidence for these enzymes playing a direct role in muscle protein breakdown (5, 6). Transcriptional regulation of these enzymes involves the FOXO1 and FOXO3a transcrip- tion factors, which are thought to regulate both the autop- hagy (7, 8) and the UPP pathways (9). These transcription factors can be inactivated by AKT, a key regulator of protein Authors' Afliations: 1 Department of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh; and 2 MRC-ARUK Centre for Muscu- loskeletal Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). I.J. Gallagher and N.A. Stephens contributed equally to the work. Corresponding Author: Kenneth C.H. Fearon, Department of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom. Phone: 44-0131-242-3615; Fax: 44-0131-242-3617; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-2133 Ó2012 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 2817 on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Upload: others

Post on 10-May-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

Human Cancer Biology

Suppression of Skeletal Muscle Turnover in CancerCachexia: Evidence from the Transcriptome in SequentialHuman Muscle Biopsies

Iain J. Gallagher1, Nathan A. Stephens1, Alisdair J. MacDonald1, Richard J.E. Skipworth1, Holger Husi1,Carolyn A. Greig1, James A. Ross1, James A. Timmons2, and Kenneth C.H. Fearon1

AbstractPurpose: Themechanisms underlyingmusclewasting in patients with cancer remain poorly understood,

and consequently there remains an unmet clinical need for new biomarkers and treatment strategies.

Experimental Design: Microarrays were used to examine the transcriptome in single biopsies from

healthy controls (n¼ 6) and in paired biopsies [pre-resection baseline (weight-loss 7%) and 8 month post-

resection follow-up (disease-free/weight-stable for previous 2 months)] from quadriceps muscle of patients

with upper gastrointestinal cancer (UGIC; n ¼ 12).

Results: Before surgery, 1,868 genes were regulated compared with follow-up (false discovery rate, 6%).

Ontology analysis showed that regulated genes belonged to both anabolic and catabolic biologic processes

withoverwhelmingdownregulation inbaseline samples.No literature-derived genes frompreclinical cancer

cachexia models showed higher expression in baseline muscle. Comparison with healthy control muscle

(n¼ 6) revealed that despite differences in the transcriptome at baseline (941 genes regulated), themuscle of

patients at follow-upwas similar to controlmuscle (2 genes regulated). Physical activity (step count per day)

did not differ between the baseline and follow-up periods (P ¼ 0.9), indicating that gene expression

differences reflected the removal of the cancer rather than altered physical activity levels. Comparative gene

expression analysis using exercise training signatures supported this interpretation.

Conclusions:Metabolic and protein turnover–related pathways are suppressed inweight-losing patients

with UGIC whereas removal of the cancer appears to facilitate a return to a healthy state, independent of

changes in the level of physical activity. Clin Cancer Res; 18(10); 2817–27. �2012 AACR.

IntroductionCancer cachexia has been defined as "a multifactorial

syndrome characterized by an ongoing loss of skeletalmuscle mass that cannot be fully reversed by conventionalnutritional support and that leads to progressive functionalimpairment" (1). Cachexia impacts negatively on the qual-ity of life of patients with cancer, response to treatment andsurvival, and thus tackling cachexia should be a centralcomponent of patient treatment. Skeletal muscle mass is

maintained by a balance between protein synthesis anddegradation, and these, in turn, are principally regulated byphysiologic inputs such as nutritional status and physicalactivity. Several systems contribute to intracellular proteinbreakdown including the ubiquitin/proteasome pathway(UPP), autophagy (lysosomal pathway), caspases, cathe-psins, and calcium-dependent calpains. Protein synthesisbegins with the association of met-tRNA and the 40Sribosome, a process regulated by elongation and initiationfactor 2 (eIF2), which, in turn, is influenced by externalsignals potentially integrated through the mTOR (2). Thebalance of activity of these processes represents the molec-ular basis for muscle mass homeostasis.

Preclinical models of muscle atrophy, which tend toreflect acute and rapid changes in muscle mass, havehighlighted the role of the UPP with increased mRNAexpression of 2 muscle-specific E3 ubiquitin ligases,MuRF-1/Trim63, and MAFbx/atrogin-1/Fbxo32 (3, 4) andevidence for these enzymes playing a direct role in muscleprotein breakdown (5, 6). Transcriptional regulation ofthese enzymes involves the FOXO1 and FOXO3a transcrip-tion factors, which are thought to regulate both the autop-hagy (7, 8) and the UPP pathways (9). These transcriptionfactors can be inactivated by AKT, a key regulator of protein

Authors' Affiliations: 1Department of Clinical and Surgical Sciences,University of Edinburgh, Edinburgh; and 2MRC-ARUK Centre for Muscu-loskeletal Ageing, College of Medical and Dental Sciences, University ofBirmingham, Edgbaston, Birmingham, United Kingdom

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

I.J. Gallagher and N.A. Stephens contributed equally to the work.

Corresponding Author: Kenneth C.H. Fearon, Department of Clinicaland Surgical Sciences, University of Edinburgh, Edinburgh EH16 4SB,United Kingdom. Phone: 44-0131-242-3615; Fax: 44-0131-242-3617;E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-2133

�2012 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org 2817

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 2: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

synthesis. Several functional studies have examined proteinsynthesis and protein degradation in rodent models ofcancer-associated muscle wasting (10–12) and these dataimply involvement of both synthesis and degradation path-ways in cancer-associated muscle atrophy in animal mod-els. Explorationof the relative contributions of anabolic andcatabolic pathways in humans is more limited and moststudies include only a small number of patients. Transcrip-tional activation of UPP components has been found in theskeletal muscle of patients with pancreatic, upper gastroin-testinal (UGI), or liver cancer (13–15) and this resulted inincreased protein or proteolytic activity in vitro (14, 16). Ithas also been reported that skeletal muscle calpain, atrogin-1, and MuRF1 mRNA remained unchanged in the skeletalmuscle of patients with gastric cancer, whereas calpainactivity was elevated even in the absence of weight loss(17). In patients with lung cancer, there was no increase inskeletal muscle mRNA expression of UPP genes but cathep-sin B mRNA was increased (18).

In contrast to the individual gene observations describedabove, we found no evidence for an increase in proteolyticgene expression, using genome-wide transcript analysis of 3types of skeletalmuscle, in a relatively large groupofweight-losing patients withUGI cancer (19), yet the same approachclearly showed that these systems were activated in humanmuscle during the more extreme situation found in theintensive care unit (20). The transcriptional analysis wasalso supported by a lack of alteration in FOXO proteinactivation; however, protein analysis suggested thatCaMKIIb activationmaymediate some aspect of suppressedprotein synthesis.

In general observations from cross-sectional studies ofcancer cachexia are impacted by patient heterogeneity,including different phases of cachexia. Nonetheless, thesetend to suggest that human cancer cachexia is not simply

"runaway" proteolysis. One approach to reduce the impactof patient heterogeneity is to study skeletal muscle changeslongitudinally. Furthermore, to determine the potentialrelevance of the tumor burden per se, determination of theskeletal muscle profile before and after removal of thetumor should be insightful. Thus, in the present study, weproduced a global transcriptional profile of human quad-riceps muscle in patients before and after potentially cura-tive surgery for UGI cancer. We show that the presence ofcancer and concurrent weight loss leads to a suppression oftranscriptional activity for genes involved in both anabo-lism and catabolism, supporting the idea that overall mus-cle turnover is suppressed in cancer cachexia. Critically, weshow that the molecular impact of tumor removal onskeletal muscle phenotype appears independent of patientphysical activity, suggesting direct cross-talk between thetumor and the process of cancer cachexia.

Materials and MethodsSubjects

A total of 18 subjectswere recruited; 6healthy control and12 patients with UGIC. Patients with UGIC had a diagnosisof esophageal (n¼ 6), gastric (n¼ 2), or pancreatic (n¼ 4)malignancy and underwent potentially curative surgery. 8patients had stage III, 2 patients had stage II, and 2 patientshad stage I disease. Five patients had completed a course ofneoadjuvant chemotherapy but had not received chemo-therapy in the 4 weeks before surgery. No subjects weretaking anabolic/catabolic agents and had uncontrolleddiabetes or known thyroid disorders. The weight-stablehealthy control comprised 5 subjects undergoing abdom-inal surgery for nonmalignant, noninflammatory condi-tions and 1 subject recruited from the community. Writteninformed consentwasobtained fromall subjects and ethicalapproval received from Lothian Research Ethics Committee(Scotland, UK). Body weight was measured with partici-pants in light clothing using a beam scale (Seca).Heightwasmeasuredusing a standardwallmountedmeasure andbodymass index (BMI)was calculated. Clinical details anddegreeof weight loss from self-reported preillness stable weightwere recorded.

Muscle biopsiesAll patients with UGIC and 5 healthy controls underwent

a baseline percutaneous quadriceps muscle biopsy follow-ing an overnight fast at the time of induction of generalanesthesia using conchotome biopsy forceps. Follow-upbiopsies for the patients with UGIC and for the healthycontrol recruited from the community were obtained fol-lowing an overnight fast under local anesthesia using aBergstrom needle with suction. Tissue samples were quicklycleaned of blood, flash-frozen in liquid nitrogen, and storedat �80�C until further analysis.

Blood measuresAll blood samples were taken following an overnight fast.

Albumin and C-reactive protein (CRP) were measured inClinical Chemistry, Royal Infirmary, Edinburgh (fully

Translational RelevanceCachexia affects 50% of all patients with cancer and

contributes to 20% of cancer-related deaths. Althoughthere have been many studies in animal models, thereare no longitudinal studies in humans to determine thepredominant alterations inmuscle. Here, we have exam-ined the transcriptional profile of human quadricepsmuscle in patients before and after potentially curativesurgery for upper gastrointestinal cancer. We show thatcancer and weight loss lead to a suppression of tran-scription for genes involved in both anabolism andcatabolism, supporting the idea that muscle turnover issuppressed in cancer cachexia. The finding that changesin the transcriptome are reversible with successfulremoval of the cancer suggests that optimal benefitmight be observed if cachexia therapy was providedduring or immediately after oncological therapy, ratherthan being left to a stage when both the cancer andcachexia are refractory to intervention.

Gallagher et al.

Clin Cancer Res; 18(10) May 15, 2012 Clinical Cancer Research2818

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 3: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

accredited by Clinical Pathology Accreditation Ltd.) usingstandard automated methods. A CRP �10 mg/L was con-sidered consistent with the presence of systemicinflammation.

Quadriceps strengthMaximum voluntary isometric quadriceps strength was

measured using an established method (21). The partici-pant was seated in an adjustable straight-backed chair withthe pelvis secured and the knee flexed at 90 degrees. A cuffwas placed around the ankle and attached via an inexten-sible chain to a strain gauge and data acquisition system(Powerlab, AD Instruments). Following instruction, theparticipant made a maximum voluntary contraction whichwas held for 5 seconds. Three separate measurements wereobtained for each limb and the highest value from thedominant limb used in subsequent analysis. Data werenormalized to body mass (N kg�1).

Physical activity monitoringPhysical activity and sedentary behavior were recorded

continuously over a 4-day period using an activPAL activitymonitor (PAL Technologies Ltd.) as described previously(22). The monitor was worn at baseline in the weekpreceding surgery and then in the week following thefollow-up assessment. The average number of steps per24 hours over the 4-day period was calculated and used foranalysis.

Molecular biologyTissue was homogenized in QIAzol (Qiagen) reagent

using a Polytron PT1200E (Kinematica AG). Total RNAwasextracted using miRNeasy columns (Qiagen) as directed bythemanufacturer with an on columnDNAse digestion step.RNA was quantified using the Nanodrop Instrument (Lab-tech Intl.). Quality and purity of RNA were examined using260/280 and 260/230 ratios. The Agilent Bioanalyzer (Agi-lent) was used to assess RNA integrity using previouslypublished protocols (19).

Reverse transcription quantitative PCRThe High Capacity RNA-to-cDNA Kit (Applied Biosys-

tems) was used to convert 500 ng of RNA to cDNAfollowing the manufacturer’s directions. Quantitative PCR(qPCR) reactions were made up to 10 mL using 5 mLPOWER SYBR-Green Master mix (Applied Biosystems),0.2 mL forward primer, 0.2 mL reverse primer, 3.6 mLH2O, and 1 mL cDNA. The final primer concentration was50 nmol/L of each primer per reaction. Reactions were runin triplicate on a StepOne Plus instrument (Applied Bio-systems). Running conditions were 95�C for 10 minutesfollowed by 40 cycles of 95�C for 15 seconds and 60�C for60 seconds. Primer efficiencies were examined with refer-ence to b-actin (which did not vary with condition—datanot shown). For genes with primer efficiencies comparablewith b-actin, we used the DDCT method for analysis. Datafrom primers with efficiencies that differed from theb-actin primers were analyzed using the standard curve

method. Primer sequences are detailed in the Supplemen-tary Material.

MicroarraysReverse transcription of RNA was carried out using the

Ovation RNA Amplification System v2 (Nugen Technolo-gies Inc.). One hundred nanograms of total RNA wasreverse-transcribed as per manufacturer’s protocol. cDNAwas purified using QIAquick PCR purification kit (Qiagen)and quantified using the Nanodrop instrument. For themicroarray study, 3.75 mg of cDNA was fragmented andlabeled with the FL-Ovation cDNA Biotin Module V2(Nugen Technologies Inc.) and hybridized to each Affyme-trix U133þ2 array (Affymetrix) using protocols provided byNugen Technologies Inc. Arrays were washed, labeled, andscanned following Affymetrix standard procedures. Rawarray data were normalized using the MAS 5.0 algorithmto a global scaling intensity of 100. Quality assurance ofarray data examined the standard quality assessmentsincluding scaling factors, glyceraldehyde–3–phosphatedehydrogenase (GAPDH) and b-actin 50/30 ratios, RLE andNUSE plots, and clustering of raw array data. Although wefound consistently high b-actin 30 to 50 ratios, an observa-tion we have made previously using the Nugen amplifica-tion protocols onmuscle-derived RNA (unpublished obser-vation), therewere noother indications that any array failedthese standard quality assurance procedures. Raw micro-array data have been deposited with Gene ExpressionOmnibus (GEO) under accession number GSE34111.

StatisticsGroup data are presented as mean and SEM. Compar-

isons were by ANOVA or Student t test as implemented in R(www.r-project.org) or SPSS (IBM). For multiple testing ofreverse transcription qPCR (RT-qPCR) data, P values werecorrected using the Bonferroni method. The raw array datawere normalized using the robust multiarray analysis(RMA) algorithm. Low variance probes were removed fromthe data set before assessment of differential regulation.Microarray data were examined using the significance anal-ysis of microarrays (SAM; ref. 23) approach and a falsediscovery rate (FDR, expected proportion of false-positives)of 10% was applied to select differentially expressed genes.Gene Ontology (GO) and Kyoto Encyclopedia and Genesand Genomes (KEGG) enrichment in differentiallyexpressed genes were examined using hypergeometric ttest as implemented in the GOstats package in R/Biocon-ductor. A tissue-enriched gene set derived from the arraydata was used as the background for this analysis tocompensate for tissue-specific GO bias (24). We also usedthe web-based Ingenuity pathway analysis (IPA) to visu-alize the gene associations modulated in the muscle tissue.IPA relies on bibilometric associations that can provideeither insight into the canonical pathways responsible for apattern of gene changes or simply a visual associationnetwork showing how the regulated genes may be inter-acting, assuming that all components are expressed in thetarget organ.

Suppressed Turnover of Skeletal Muscle in Cancer Cachexia

www.aacrjournals.org Clin Cancer Res; 18(10) May 15, 2012 2819

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 4: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

ResultsDemographics

Demographic details for the groups are shown in Table 1.Age was not significantly different between patients withUGIC and healthy controls. BMI and weight in the cancergroup did not differ significantly from the control group atany time point. The median time between baseline andfollow-up biopsies was 8months (range, 5–12months). Allpatients with UGICwere clinically cancer-free at the time ofrepeat biopsy. Mean weight loss from self-reported usualweight was 7.4% at baseline and increased to 13.8% atfollow-up. However, this did not occur in a linear fashionbut rather was more rapid immediately following surgerybefore stabilizing in the 2 months before follow-up biopsy(Fig. 1A). Eight of the 12 patients with UGIC were includedin the activity meter study and neither age and BMI norweight differed significantly from the overall UGIC group(data not shown). In patients with UGIC, there was anincrease in quadriceps strength between baseline and fol-low-up of 15%, but this did not reach statistical significance(P ¼ 0.2; Table 1; Fig. 1C). Overall physical activity (stepcount) remained stable between baseline and follow-up(Table 1; Fig. 1B).

Albumin and CRPAt baseline, patients with UGIC had reduced albumin

levels compared with healthy controls [36.0 (0.5) vs. 42.4(1.3) g/L, P < 0.001; Table 1). At the time of follow-upbiopsy, albumin had returned to within the reference range(36–44 g/L; Table 1). CRP did not differ between baselineand follow-up (9.75 vs. 6.0 mg/L, P ¼ 0.5) and was notsignificantly different compared with healthy controls ateither time point (Table 1).

Paired muscle biopsy transcriptomeUsing paired samples from the patients with UGIC (n ¼

12) obtained immediately preoperatively (baseline: cancerpresent; weight-losing) and at a median of 8 months post-operatively (follow-up: cancer resected; weight-stable for atleast last 2 months), we examined global gene expressionusing microarrays. Paired analysis within the SAM method(23) identified 1,868 regulated genes with an FDR of 6%and a fold change of at least 30%, 1,747 downregulated and121upregulated in baseline samples comparedwith follow-up samples (Fig. 2A). We validated expression of selectedregulated genes by RT-qPCR and found good agreementbetween the array and RT-qPCR data (Table 2).

To examine the biologic context of the regulated genes,we used the hypergeometric t test to identify enriched GeneOntology Biological Processes (GOBP) and KEGG path-ways. Using a background list of genes detectably expressedin muscle tissue (24), we identified cellular metabolicprocess as the most enriched biologic process (P <0.001). The enriched GOBP categories included severalbroadmetabolic processes (e.g., metabolic process, primarymetabolic process), muscle growth categories (e.g., striatedmuscle hypertrophy, regulation of muscle hypertrophy),catabolism categories (e.g., endoplasmic reticulum–associ-ated protein catabolic process, ubiquitin-dependent pro-tein catabolic process), and anabolism categories (e.g.,response to insulin stimulus, response to protein stimulus).In each of these categories, the genes involved were over-whelmingly downregulated in the baseline samples. Select-ed categories are presented in Table 3 and full enrichmentresults are available in the Supplementary Material.Enriched KEGG pathways included the calcium signalingpathway, the peroxisome pathway, and EGF receptor sig-naling (ErbB signaling pathway).

Table 1. Demographic data for paired samples (baseline and follow-up) and healthy control subjects in thestudy

Paired samples

Baseline (n ¼ 12) Follow-up 2 (n ¼ 12) Controls (n ¼ 6)

Gender (M/F) 10/2 10/2 4/2Age 65 65 58BMI, kg/m2 26.4 (1.3) 24.6 (1.3)a 28.2 (1.7)Weight, kg 78.2 (4.5) 72.8 (4.4) 84.9 (7.2)Change from usual weight (%) �7.3 (2.7)b �13.8 (2.7)a,b 0CRP, mg/L 9.8 (3.6) 6.0 (3.4) 1.8 (1.0)Albumin, g/L 36 (0.5)b 42 (0.9)a 42 (1.3)Step count/24 h (n ¼ 8 per group) 5,607 (1,210) 5,675 (917) NAQuadriceps strength, N kg�1 3.5 (0.3) 4.5 (0.9) NA

NOTE: Data (except gender split and age) are presented as mean (SEM).Abbreviation: F, female; M, male; N, Newton.aSignificantly different from baseline (P < 0.01, paired t test).bSignificantly different from the controls (P < 0.01).

Gallagher et al.

Clin Cancer Res; 18(10) May 15, 2012 Clinical Cancer Research2820

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 5: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

We used IPA to examine a subset of 350 robustly regu-lated genes that did not overlap with either endurance (25)or resistance training (26) data sets. Network analysis,relying on the IPA database revealed the proteasome, NF-kB, and caspases as potential network hubs connected tomany downregulated transcripts (Fig. 2B). Downregulationof these network genes suggests that activation of the hubgenes is lower preoperatively supporting the lack of activa-tion of proteolytic genes in the differential expressionanalysis. Using gene set enrichment analysis and the onlineoPOSSUM tool, we examined the gene expression data forgenes under the control of specific transcription factors andfound no significant results, suggesting that no singlecanonical signaling pathway is driving the muscle cachexiaprofile.

Preclinicalmodels (3, 4, 27) have identified a set of genes,commonly termed atrogenes, which are involved in cancer-driven muscle wasting. In particular, the levels of the E3ligases atrogin-1 (FBXO32) and MuRF1 (TRIM63) are ele-vated in the muscle of cachectic animals. These genes werenot differentially expressed in the microarray data, norwhen examined by RT-qPCR (atrogin-1 P ¼ 1; MuRF1 P¼0.69; paired t test; Table 4). Examinationof the autophagygenes BNIP3 and GABARAPL1 (28, 29) also revealed nochanges in expression of these genes between baseline,follow-up, or healthy control groups (Table 4). Glucocorti-coids play a role inmuscle wasting in some conditions (30).We found no differences in the mRNA levels of the gluco-corticoid receptor (NR3C1) and 11b-HSD between thegroups (Table 4).

Physical activityTo control for potential changes in physical activity in the

baseline versus follow-upperiods, the average step count per24 hours was measured (n ¼ 8 of 12 patients with UGIC).Mean (SEM) step count/24 hours at baseline was 5,607(1,210) and 5,675 (917) at follow-up. This difference wasnot significant (P ¼ 0.9; paired t test; Fig. 1B). As a com-plementary approach to direct measurement of physicalactivity, we also compared our list of differentially expressedgeneswith those identified recently in the response to2 typesof exercise training (25, 26). Of the 848 endurance trainingresponsive genes, only 60 overlapped with those regulatedbetween the baseline and follow-up time points (see Sup-plementary Material) and even fewer overlapped withstrength training. To compare the response in cancer cachex-ia with that seen in dietary restriction, we examined theoverlap with the 2,839 differentially expressed genes iden-tified in response to simple dieting (31). Only 12 genesoverlapped between our data set and those regulated inskeletal muscle in response to simple dieting. To examineany relationship with the ageing process, we compared ourdata set with a large study of young versus elderly muscle(manuscript submitted) and found no association betweenthedata sets. Thus, usingmultiple array analysis strategieswecan conclude that the major phenotype of skeletal muscle"responding" to removal of a tumor does not appear relatedto changes in physical activity, nutritional status, or age.

Figure 1. Weight changes (A), mean step counts (B), and normalizedquadriceps strength (C) from baseline to follow-up time points. A, weightwas significantly decreased from reported usual weight at baseline andboth follow-up time points (�, P < 0.05). At both follow-up time points,weight was lower than baseline (†,P < 0.05) but did not differ between thefollow-up time points. B, mean step count over 4 days was not differentbetween the baseline and follow-up time points. C, body massnormalized quadriceps strength (N kg�1) improved by 15% between thebaseline and follow-up time points, although this did not reach statisticalsignificance. FU, follow-up.

Suppressed Turnover of Skeletal Muscle in Cancer Cachexia

www.aacrjournals.org Clin Cancer Res; 18(10) May 15, 2012 2821

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 6: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

Comparison with healthy control muscleComparison of the muscle from the patients with UGIC

at both the baseline and follow-up time points with musclefrom weight-stable healthy control subjects revealed 941genes with lower expression (FDR < 10%) in baseline UGICmuscle compared with healthy control muscle. At UGICbaseline, no genes were expressed at higher levels than incontrol muscle with FDR < 10%. Of the 941 downregulatedgenes, 558 were also regulated between the baseline andfollow-up samples and all except one (TFDP2, transcriptionfactor Dp-2) were higher at follow-up. Highly enrichedGOBP categories included lipid oxidation, catabolic pro-cess, protein polyubiquitination, muscle structure develop-ment, and striated muscle hypertrophy. Enriched KEGGpathways included fatty acid metabolism, metabolic path-ways, peroxisome, oxidative phosphorylation, ubiquitin-mediated proteolysis, and regulation of autophagy (seeSupplementary Material). Comparison of gene expressionbetween healthy control muscle and the follow-up samplesrevealed only 2 genes differentially expressed within the

FDR cutoff point (NR1D2 and HDAC9). Thus, the follow-up samples had a transcriptomic profile indistinguishablefrom healthy muscle, albeit that the total sample size waslimited for this unpaired analysis.

DiscussionCachexia is an important cancer co-morbidity impacting

onquality of life and response to treatment. In this study,weexamined the transcriptomic response of skeletal musclebefore and after potentially curative surgery for UGIC.Resection of UGIC is routinely associated with a weightloss of 7% to 15% of body weight (32) and patients willgenerally not regain nor return to their preoperative weight.Weight loss during the postsurgical phase is considered tobe largely due to loss of fat mass with only limited changesin muscle mass (32), suggesting that greater emphasisshould be placed on the restoration of muscle phenotypeand function rather than body weight. The present studyinvestigated patients with UGIC undergoing tumor

Figure 2. Heatmap representation (A) and IPAderived network (B) of the genes differentially expressed between the baseline and follow-up time points. A, geneexpression is shown for the baseline (B; green bar) and follow-up (FU; blue bar) time points as well as in the healthy control group (HC; pink bar). Colorsrepresent scaled and centered expression values: red represents higher expression, blue represents lower expression. B, IPA was used to derive a networkfrom 350 genes differentially expressed between baseline and follow-up and not shared by training or dieting responses. All genes in the network weredownregulated at baseline.

Gallagher et al.

Clin Cancer Res; 18(10) May 15, 2012 Clinical Cancer Research2822

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 7: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

resection with an average weight loss of 7% body weight(i.e., fulfilling the diagnostic criteria for cancer cachexia;ref. 1). Patients underwent a preoperative baseline quadri-ceps biopsy with a repeat quadriceps biopsy conductedapproximately 8 months later when all patients were clin-ically disease-free and had been weight-stable for at least 2months (Table 1; Fig. 1A). There is a marked decline inphysical activity immediately following major abdominalsurgery. However, by 8 months after surgery, patients’quadriceps strength tended to be higher than baseline andoverall physical activity had returned to baseline levels. It isthus reasonable to consider that the patients in the presentstudy had recovered from the net catabolic effects of boththeir cancer cachexia and surgery. Sequential paired biop-sies allowed analysis of changes in skeletal muscle pheno-

type during recovery whereas comparison with a group ofweight-stable healthy controls allowed determination ofthe completeness of recovery. In the present study, patientswith UGIC showed normalization of muscle phenotypepostsurgery (Table 1; Fig. 2A). Strikingly neither the cancercachexia signature nor the recovery of healthy muscle phe-notype appeared to be related to levels of physical activity.

The present study shows that in human skeletal muscle,of the 1,868 regulated genes associated with cancer andweight loss, the vast majority (94%) are downregulated.Category analysis of the differentially expressed genesshowed that both anabolic (e.g., muscle organ develop-ment, response to insulin stimulus) and catabolic (e.g.,ubiquitin-dependent protein catabolic process, regulationof proteasomal protein catabolic process) process gene

Table 2. RT-qPCR validation of microarray data

Baseline vs. follow-up Healthy vs. baseline

Array (FDR) qPCR (P) Array (FDR) qPCR (P)

COMP 3.91 (4.1) 4.3 (<0.01) 0.7 (53) 0.36 (0.02)ADIPOQ 3.3 (4.1) 3.2 (<0.01) 0.44 (48) 0.45 (0.06)MMP3 3.2 (6.1) 2.7 (0.04) 0.66 (53) 0.59 (0.2)PCK1 3.0 (6.1) 2.5 (0.01) 0.57 (50) 0.52 (0.14)ANGPTL7 2.8 (6.1) 2.5 (<0.01) 0.63 (51) 0.62 (0.46)HSP90AB1 0.45 (6.1) 0.5 (0.04) 2.2 (10) 2.3 (0.10)SLC25A37 0.45 (6.1) 0.5 (<0.01) 2.6 (5.1) 2.6 (<0.01)PROX1 0.45 (2.1) 0.5 (<0.01) 2.8 (0.0) 2.6 (<0.01)RCAN1 0.44 (0.0) 0.5 (<0.01) 2.0 (5.5) 2.4 (<0.01)HINT3 0.25 (0.0) 0.2 (<0.01) 3.1 (0.0) 3.1 (<0.01)

NOTE: Array data are presented as fold change (FDR expressed as a percentage) and RT-qPCR data as fold change (Bonferronicorrected P value for difference in means). N¼ 12 paired samples for follow-up versus baseline analysis; N¼ 5 healthy control (for onesample there was insufficient RNA for RT) and 12 baseline samples for healthy versus baseline analysis. RT-qPCR data were analyzedby paired t tests for follow-up versus baseline analysis and by unpaired t tests for healthy control versus baseline analysis.

Table 3. GOBP categories regulated between baseline and follow-up time points

GOBP ID PExpectedcount

Actualcount

Annotatedgenes on array Term

GO:0044237 0.00 784 859 5,042 Cellular metabolic processGO:0006350 0.01 288 320 1,854 TranscriptionGO:0045449 0.00 277 315 1,784 Regulation of transcriptionGO:0009056 0.03 133 153 858 Catabolic processGO:0030163 0.01 45 59 288 Protein catabolic processGO:0006511 0.01 34 48 216 Ubiquitin-dependent protein

catabolic processGO:0032868 0.02 15 24 99 Response to insulin stimulusGO:0051789 0.03 13 20 85 Response to protein stimulusGO:0061061 0.04 37 47 236 Muscle structure developmentGO:0007517 0.01 31 43 197 Muscle organ developmentGO:0060537 0.04 20 28 131 Muscle tissue developmentGO:0014706 0.02 19 28 124 Striated muscle tissue development

Suppressed Turnover of Skeletal Muscle in Cancer Cachexia

www.aacrjournals.org Clin Cancer Res; 18(10) May 15, 2012 2823

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 8: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

expression was suppressed. Furthermore, there was a lack ofsubstantive overlap with transcriptomic signatures fromendurance training, strength training, ageing, or simpledieting (25, 26, 31, 33). This is perhaps not surprising aseach of these situationsmay be considered within the rangeof normal physiology rather thanpathologic states.Wewereable to identify the inflammatory transcription factorNF-kBas a hub gene using IPA, however, this gene is linked tomany aspects of muscle remodeling including responses toendurance training (25). Other approaches to identifytranscription factors [a priori gene set enrichment analysis(GSEA) and post hoc analysis with the web-based oPOSSUMsoftware] did not yield any significant results, suggestingthat no single transcription factor network is involved inhuman cancer cachexia. Limited numbers, variable humandata, and transcription factor redundancy are factors thatmight constrain the discovery of such underlying controlgenes.

Comparison of the patients with UGIC with weight-stable noncancer healthy controls shows that followingremoval of cancer and stabilization of weight loss, thetranscriptome essentially returns tonormal. Taken together,wewould hypothesize thatmuscle turnover is suppressed inthe early stages of cancer cachexia, butwith successful cancertreatment, these changes are reversible and may directlyrelate to the presence of the tumor. Our observations mayalso have implications for the timing of interventions suchas physical therapy, as it may be more productive to inter-vene during this stable period when the molecular pheno-type is apparently healthy and function improving. Alter-natively, the recovery of the molecular phenotype could beaccelerated by earlier interventions and future studies maybe able to address this point.

In a recent study on the response of adipose tissue tohuman cancer cachexia, 364 genes were downregulated and61 genes upregulated in abdominal subcutaneous whiteadipose tissue (34). Similar to the present study, pathwayanalysis indicated that downregulated genes were involvedin cytoskeleton and extracellular matrix processes. Changesin gene expression were reciprocal to those observed inobesity, suggesting that regulation of fat mass in cachexiamay be dominated by the effects of reduced food intake. Inthe present study, the gene changes in muscle were dissim-

ilar to those seen in simple dieting (31), thereby confirmingthe essential phenotype of cachexia in that the response ofmuscle to cancer is distinct from simple starvation (1, 35)and cannot be explained by cancer-induced anorexia alone.This observation may also explain the suboptimal skeletalmuscle response to nutritional supplementation observedin patients with cancer.

The transcriptomic signal that we detected at baseline inpatients with UGIC suggests depression of muscle turnoverin patients with cancer-associated weight loss supportingearlier ideas that both global protein synthesis and break-down were suppressed. While muscle turnover could be afunction of changes in physical activity, we did not observeany differences in 24-hour step count between baseline andfollow-up assessments and found a biologically relevantimprovement of 15% in normalized quadriceps strengthafter tumor removal. Furthermore, the genes differentiallyexpressed after removal of the tumor showed little overlapwith genes expressed after exercise training, supporting theconclusion that restoration of muscle phenotype was notsimply a retraining response. The median time intervalbetween operation and follow-up biopsy was 8 monthsandmuscle function is generally considered to be recoveredby this time (36).

The loss of skeletal muscle protein in cancer cachexia hasbeen attributed to both alterations in the transcription ofspecific genes leading to changes inmuscle turnover and/ora global reduction in cellular RNA reducing overall trans-lational capacity. A global reduction in RNA abundancemight be due to reduced transcriptional capacity as a resultof loss of myonuclei secondary to enhanced apoptosis orreduced satellite cell recruitment. We have previouslyobserved evidence of apoptosis in cachectic patients withcancer similar to those involved in the present study (37).However, Lundholm and colleagues reported that humanskeletalmuscle RNA content is unaffected by the presence ofa tumor, whereas in animal models, there was a decrease inRNA content (38). While animal models carrying a varietyof tumors have shown reduced RNA content inmuscle (39–41) in 2 of these models, there was also evidence forconcurrent increases in mRNA expression of proteasomalsubunits (15, 40). Notably, we have found a reduction inmRNA expression for proteasomal pathway components in

Table 4. RT-qPCR data for selected atrophy-associated genes

Baseline vs. follow-up Baseline vs. control Follow-up vs. control

TRIM63 (MuRF1) 0.9 (1.00) 1.2 (0.68) 1.3 (0.22)FBXO32 (Atrogin-1) 1.0 (1.00) 1.0 (1.00) 1.0 (1.00)BNIP3 0.9 (0.18) 0.7 (0.22) 0.8 (0.50)GABARAPL1 0.8 (0.26) 0.7 (0.14) 0.8 (0.70)NR3C1 0.9 (0.78) 0.8 (0.16) 0.8 (0.36)HSD11b1 1.1 (0.36) 1.5 (0.12) 1.3 (0.06)

NOTE: Data are shown as fold change (Bonferroni corrected P value for difference between means). Baseline versus follow-up datawere analyzed using paired t tests.

Gallagher et al.

Clin Cancer Res; 18(10) May 15, 2012 Clinical Cancer Research2824

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 9: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

human UGIC in this study and no evidence for changes inmRNA for these components previously (19).Direct evidence for the involvement of specific proteolytic

and synthetic pathways in human cancer cachexia is limit-ed. However, the balance of evidence, based on stableisotope and global transcript analysis, including the presentstudy, appears to favor the conclusion that suppression ofboth processes occurs, with the reduction in synthetic path-ways exceeding suppression of the proteolytic pathways. Inline with this, protein degradation as measured by netrelease of 3-methylhistidine across the leg was lower inpatients with cancer than in healthy controls or acutely illpatients and accompanied by anabolic blunting in responseto feeding, suggesting a net catabolic status rather thanincreased degradation (42). Others have also reporteddepressed skeletal muscle protein synthesis in the face ofmaintenance of protein breakdown, again suggesting rela-tive catabolic advantage (43).What remains to be determined is the precise nature of

the cross-talk between the tumor and the process of skeletalmuscle cancer cachexia. Certain transcript changes in thepresent study might be explored as biomarkers of cancercachexia and provide insight into the nature of the processor an early indication of patient responsiveness to anintervention.We found increased serum cartilage oligomer-ic matrix protein (COMP) gene expression in cachecticmuscle and a similar pattern of expression has been seenin muscle damage in marathon runners (44). Similarly,expression of matrix metalloproteinase 3 (MMP3), alsoincreased in cachectic muscle, is increased in models ofmuscle injury (45). These findings are consistent with oursuggestion that removal of cancer allows skeletal muscle torecover from net catabolism. We have recently publisheddatadetailing a role forADIPOQ,upregulated at baseline, asa network hub in cachexia (46) suggesting that this mighthave a role in cancer-associated muscle atrophy or be asensitive integrator of the net underlying processes (e.g.,metabolic status of the muscle). Increased expression of thephosphoenolpyruvate carboxykinase 1 (PCK1) gene hasbeen found in lean rodents (47) in which it has been linkedto increased endurance capacity. However, our finding ofincreased PCK1 expression in cachectic muscle may suggestthat this is representative of a possible compensatoryresponse in the face of catabolic advantage.Of the genes downregulated at baseline, regulator of

calcineurin 1 (RCAN1) inhibits calcineurin signaling andis upregulated in muscle adapting to eccentric exercise (48)and calcineurin activation may be a key feature of muscleadaptation to endurance exercise in humans (25). Againthese changes suggest that the muscle is attempting tocompensate for the influence of the tumor, supporting ourconclusion that muscle remodeling/turnover is specificallysuppressed by the presence of cancer. In rodents, inactiva-tion of the transcription factor prospero homeobox 1(PROX1) leads to disorganization of cardiac muscle (49).If this function was recapitulated in skeletal muscle, thenreducedPROX1 expression could indicate a failure of propermuscle remodeling. Our previous analysis of gene expres-

sion in muscles of patients with cancer showed that expres-sion of the exercise-activated genes CAMk2b and TIE1 wasincreased in patients with increasing weight loss (19).However, although at the follow-up time point, patientswith UGIC were weight-stable from the perspective of thepostoperative period, their overall weight was still lowerthan at the point of diagnosis. Therefore, it is possible thatCAMk2b and TIE1 upregulation are indicators of musclemass regulation, an idea consistent with the relatively clearability of these genes to identify evenmodest cancer cachex-ia muscle changes (19). CAMk2b and TIE1 expression hasbeen shown to be activated in multiple muscle groups,across clinical centers and the association with weight lossin this new longitudinal study suggests they are "longerterm" markers of muscle status and thus potential biomar-kers for weight loss in cancer cachexia. Nevertheless, theoptimal biomarker would predict the potential for induc-tion of cancer cachexia in the individual and to establishand validate such a biomarker would require much largerstudies.

Current therapeutic approaches to the management ofmuscle wasting in cancer cachexia include such generalmeasures as nutritional support and exercise interventions(50). Difficulties with low compliance and negligible ben-efit have stimulatedmuch interest in measures to overcomehypoanabolism or reducing increased catabolism. The pre-dominance of an activated UPP driving protein breakdownin some acute rodent models of muscle atrophy has ledmany to suggest that the UPP should be targeted in patientswith cancer cachexia. While we cannot discount the rele-vance of the UPP at some point during the development ofcancer cachexia (e.g., during the acute phase of a period ofprolonged bed rest), the findings of the present studysuggest that more emphasis should be given to the stimu-lation of anabolism and, in particular, blocking of thespecific effect that tumor burden has on muscle signaling.The finding that changes in the transcriptome are reversiblewith successful removal of the cancer suggests that optimalbenefit might be observed if such cachexia therapy wasprovided during or immediately after oncological therapy,rather than being left to a stage when both the cancer andcachexia are refractory to intervention (1).

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Grant SupportThis project was funded by an award to K.C.H. Fearon, C.A. Greig, J.A.

Ross, and J.A. Timmons from the Translational Medicine Research Collab-oration–a consortium of the Universities of Aberdeen, Dundee, Edinburgh,and Glasgow, the four associated Health Boards (Grampian, Tayside,Lothian, and Greater Glasgow and Clyde), Scottish Enterprise, and WyethPharmaceuticals. Additional funding was obtained from CRUK (K.C.H.Fearon), Capacity Building Grant (SUPAC) from the NCRI (K.C.H. Fearon),and Aileen Lynn Bequest Fund (A.J. MacDonald).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received August 18, 2011; revised January 27, 2012; accepted February 10,2012; published OnlineFirst March 27, 2012.

Suppressed Turnover of Skeletal Muscle in Cancer Cachexia

www.aacrjournals.org Clin Cancer Res; 18(10) May 15, 2012 2825

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 10: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

References1. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL,

et al. Definition and classification of cancer cachexia: an internationalconsensus. Lancet Oncol 2011;12:489–95.

2. Fingar DC, Richardson CJ, Tee AR, Cheatham L, Tsou C, Blenis J.mTOR controls cell cycle progression through its cell growth effectorsS6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol CellBiol 2004;24:200–16.

3. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, et al.Identification of ubiquitin ligases required for skeletal muscle atrophy.Science 2001;294:1704–8.

4. GomesMD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1, amuscle-specific F-box protein highly expressed during muscle atro-phy. Proc Natl Acad Sci U S A 2001;98:14440–5.

5. Lagirand-Cantaloube J, Cornille K, Csibi A, Batonnet-Pichon S, Lei-bovitch MP, Leibovitch SA. Inhibition of atrogin-1/MAFbx mediatedMyoD proteolysis prevents skeletal muscle atrophy in vivo. PLoS One2009;4:e4973.

6. CohenS, Brault JJ, Gygi SP,GlassDJ, Valenzuela DM,Gartner C, et al.During muscle atrophy, thick, but not thin, filament components aredegraded by MuRF1-dependent ubiquitylation. J. Cell Biol 2009;185:1083–95.

7. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del PiccoloP, et al. FoxO3 controls autophagy in skeletal muscle in vivo. CellMetab 2007;6:458–71.

8. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, et al. FoxO3coordinately activates protein degradation by the autophagic/lyso-somal and proteasomal pathways in atrophying muscle cells. CellMetab 2007;6:472–83.

9. Sacheck JM,Hyatt J-PK, Raffaello A, Jagoe RT, RoyRR, Edgerton VR,et al. Rapid disuse and denervation atrophy involve transcriptionalchanges similar to those of muscle wasting during systemic diseases.FASEB J 2007;21:140–55.

10. Beck SA, Smith KL, Tisdale MJ. Anticachectic and antitumor effect ofeicosapentaenoic acid and its effect on protein turnover. Cancer Res1991;51:6089–93.

11. Smith KL, Tisdale MJ. Increased protein degradation and decreasedprotein synthesis in skeletal muscle during cancer cachexia. Br JCancer 1993;67:680–5.

12. Samuels SE, Knowles AL, Tilignac T, Debiton E, Madelmont JC, AttaixD. Protein metabolism in the small intestine during cancer cachexiaand chemotherapy in mice. Cancer Res 2000;60:4968–74.

13. WilliamsA, SunX, Fischer JE, HasselgrenPO. The expression of genesin the ubiquitin-proteasome proteolytic pathway is increased in skel-etal muscle from patients with cancer. Surgery 1999;126:744–9; dis-cussion 749–50.

14. Bossola M, Muscaritoli M, Costelli P, Grieco G, Bonelli G, Pacelli F,et al. Increased muscle proteasome activity correlates with diseaseseverity in gastric cancer patients. Ann Surg 2003;237:384–9.

15. Khal J, Wyke SM, Russell ST, Hine AV, Tisdale MJ. Expression of theubiquitin-proteasome pathway and muscle loss in experimental can-cer cachexia. Br J Cancer 2005;93:774–80.

16. Khal J, Hine AV, Fearon KCH, Dejong CHC, Tisdale MJ. Increasedexpression of proteasome subunits in skeletal muscle of cancerpatients with weight loss. Int J Biochem Cell Biol 2005;37:2196–206.

17. Smith IJ, Aversa Z, Hasselgren P-O, Pacelli F, Rosa F, Doglietto GB,et al. Calpain activity is increased in skeletal muscle from gastriccancer patients with no orminimal weight loss. Muscle Nerve 2011;43:410–4.

18. Jagoe R, Redfern CPF, Roberts RG, Gibson GJ, Goodship THJ.Skeletal muscle mRNA levels for cathepsin B, but not componentsof the ubiquitin-proteasome pathway, are increased in patients withlung cancer referred for thoracotomy. Clin Sci (Lond) 2002;102:353–61.

19. Stephens NA, Gallagher IJ, Rooyackers O, Skipworth RJ, Tan BH,Marstrand T, et al. Using transcriptomics to identify and validate novelbiomarkers of human skeletal muscle cancer cachexia. Genome Med2010;2:1.

20. FredrikssonK, Tj€ader I, Keller P, PetrovicN, AhlmanB, Sch�eeleC, et al.Dysregulation of mitochondrial dynamics and the muscle transcrip-

tome in ICU patients suffering from sepsis induced multiple organfailure. PLoS One 2008;3:e3686.

21. EdwardsRH,YoungA,HoskingGP, JonesDA.Human skeletalmusclefunction: description of tests and normal values. Clin Sci Mol Med1977;52:283–90.

22. Dahele M, Skipworth RJE, Wall L, Voss A, Preston T, Fearon KCH.Objective physical activity and self-reported quality of life in patientsreceiving palliative chemotherapy. J Pain Symptom Manage 2007;33:676–85.

23. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarraysapplied to the ionizing radiation response. Proc Natl Acad Sci U S A2001;98:5116–21.

24. Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP,et al. Integration of microRNA changes in vivo identifies novel molec-ular features of muscle insulin resistance in Type 2 diabetes. GenomeMed 2010;2:9.

25. Keller P, Vollaard NBJ, Gustafsson T, Gallagher IJ, Sundberg CJ,Rankinen T, et al. A transcriptional map of the impact of enduranceexercise training on skeletal muscle phenotype. J Appl Physiol2011;110:46–59.

26. Melov S, Tarnopolsky MA, Beckman K, Felkey K, Hubbard A. Resis-tance exercise reverses aging in human skeletal muscle. PLoS One2007;2:e465.

27. Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, et al.Multiple types of skeletal muscle atrophy involve a common programof changes in gene expression. FASEB J 2004;18:39–51.

28. Mammucari C, Schiaffino S, Sandri M. Downstream of Akt: FoxO3 andmTOR in the regulation of autophagy in skeletal muscle. Autophagy2008;4:524–6.

29. Zhao J, Brault JJ, Schild A, Goldberg AL. Coordinate activation ofautophagy and the proteasome pathway by foxO transcription factor.Autophagy 2008;4:378–80.

30. Schakman O, Gilson H, Thissen JP. Mechanisms of glucocorticoid-induced myopathy. J Endocrinol 2008;197:1–10.

31. Larrouy D, Barbe P, Valle C, D�ejean S, Pelloux V, Thalamas C, et al.Gene expression profiling of human skeletal muscle in response tostabilized weight loss. Am J Clin Nutr 2008;88:125–32.

32. Liedman B, Andersson H, Bosaeus I, Hugosson I, Lundell L. Changesin body composition after gastrectomy: results of a controlled, pro-spective clinical trial. World J Surg 1997;21:416–20; discussion 420–1.

33. Timmons JA, Jansson E, Fischer H, Gustafsson T, Greenhaff PL,Ridden J, et al. Modulation of extracellular matrix genes reflects themagnitude of physiological adaptation to aerobic exercise training inhumans. BMC Biol 2005;3:19.

34. Dahlman I,Mejhert N, Linder K, Agustsson T,MutchDM,KulyteA, et al.Adipose tissuepathways involved inweight loss of cancer cachexia. BrJ Cancer 2010;102:1541–8.

35. NixonDW, LawsonDH. Nutritional support of the cancer patient. HospFormul 1983;18:616–9.

36. Mathur S, Plank LD, Hill AG, Rice MA, Hill GL. Changes in bodycomposition, muscle function and energy expenditure after radicalcystectomy. BJU Int 2008;101:973–7; discussion 977.

37. Busquets S, Deans C, Figueras M, Moore-Carrasco R, L�opez-Sor-iano FJ, Fearon KCH, et al. Apoptosis is present in skeletal muscle ofcachectic gastro-intestinal cancer patients. Clin Nutr 2007;26:614–8.

38. LundholmK, Edstr€omS, Ekman L, Karlberg I, Bylund AC, Scherst�en T.A comparative study of the influence of malignant tumor on hostmetabolism in mice and man: evaluation of an experimental model.Cancer 1978;42:453–61.

39. Emery PW, Lovell L, Rennie MJ. Protein synthesis measured in vivo inmuscle and liver of cachectic tumor-bearing mice. Cancer Res1984;44:2779–84.

40. Baracos VE, DeVivo C, Hoyle DH, Goldberg AL. Activation of the ATP-ubiquitin-proteasome pathway in skeletal muscle of cachectic ratsbearing a hepatoma. Am J Physiol 1995;268:E996–1006.

41. Bhogal AS, Lorite ML, TisdaleMJ. Changes in nucleic acid and proteinlevels in atrophying skeletalmuscle in cancer cachexia. AnticancerRes2006;26:4149–54.

Gallagher et al.

Clin Cancer Res; 18(10) May 15, 2012 Clinical Cancer Research2826

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 11: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

42. Lundholm K, Bennega�rd K, Ed�en E, Svaninger G, Emery PW,

Rennie MJ. Efflux of 3-methylhistidine from the leg in cancerpatients who experience weight loss. Cancer Res 1982;42:4807–11.

43. Dworzak F, Ferrari P, Gavazzi C, Maiorana C, Bozzetti F. Effects ofcachexia due to cancer on whole body and skeletal muscle proteinturnover. Cancer 1998;82:42–8.

44. KimHJ, LeeYH, KimCK.Changes in serumcartilage oligomericmatrixprotein (COMP), plasmaCPKandplasma hs-CRP in relation to runningdistance in a marathon (42.195 km) and an ultra-marathon (200 km)race. Eur J Appl Physiol 2009;105:765–70.

45. Goetsch SC, Hawke TJ, Gallardo TD, Richardson JA, Garry DJ.Transcriptional profiling and regulation of the extracellular matrixduring muscle regeneration. Physiol Genomics 2003;14:261–71.

46. Tan BHL, Ross JA, Kaasa S, Skorpen F, Fearon KCH. Identification ofpossible genetic polymorphisms involved in cancer cachexia: a sys-tematic review. J Genet 2011;90:165–77.

47. Novak CM, Escande C, Gerber SM, Chini EN, Zhang M, Britton SL,et al. Endurance capacity, not body size, determines physical activitylevels: role of skeletal muscle PEPCK. PLoS One 2009;4:e5869.

48. Mahoney DJ, Safdar A, Parise G,Melov S, FuM,MacNeil L, et al. Geneexpression profiling in human skeletal muscle during recovery fromeccentric exercise. Am J Physiol Regul Integr Comp Physiol 2008;294:R1901–10.

49. Risebro CA, Searles RG, Melville AAD, Ehler E, Jina N, Shah S, et al.Prox1 maintains muscle structure and growth in the developing heart.Development 2009;136:495–505.

50. Fearon KCH. Cancer cachexia: developing multimodal therapy for amultidimensional problem. Eur J Cancer 2008;44:1124–32.

Suppressed Turnover of Skeletal Muscle in Cancer Cachexia

www.aacrjournals.org Clin Cancer Res; 18(10) May 15, 2012 2827

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133

Page 12: Suppression of Skeletal Muscle Turnover in Cancer Cachexia ... · begins with the association of met-tRNA and the 40S ribosome, a process regulated by elongation and initiation factor

2012;18:2817-2827. Published OnlineFirst March 27, 2012.Clin Cancer Res   Iain J. Gallagher, Nathan A. Stephens, Alisdair J. MacDonald, et al.   BiopsiesEvidence from the Transcriptome in Sequential Human Muscle Suppression of Skeletal Muscle Turnover in Cancer Cachexia:

  Updated version

  10.1158/1078-0432.CCR-11-2133doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2012/05/10/1078-0432.CCR-11-2133.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/18/10/2817.full#ref-list-1

This article cites 50 articles, 13 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/18/10/2817.full#related-urls

This article has been cited by 5 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/18/10/2817To request permission to re-use all or part of this article, use this link

on May 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 27, 2012; DOI: 10.1158/1078-0432.CCR-11-2133