Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from...

12
Integrated Systems and Technologies Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen Heitzer 1 , Martina Auer 1 , Christin Gasch 6 , Martin Pichler 3 , Peter Ulz 1 , Eva Maria Hoffmann 1 , Sigurd Lax 5 , Julie Waldispuehl-Geigl 1 , Oliver Mauermann 6 , Carolin Lackner 2 , Gerald Hoer 2 , Florian Eisner 3 , Heinz Sill 4 , Hellmut Samonigg 3 , Klaus Pantel 6 , Sabine Riethdorf 6 , Thomas Bauernhofer 3 , Jochen B. Geigl 1 , and Michael R. Speicher 1 Abstract Circulating tumor cells (CTC) released into blood from primary cancers and metastases reect the current status of tumor genotypes, which are prone to changes. Here, we conducted the rst comprehensive genomic proling of CTCs using arraycomparative genomic hybridization (CGH) and next-generation sequencing. We used the U.S. Food and Drug Administrationcleared CellSearch system, which detected CTCs in 21 of 37 patients (range, 1202/7.5 mL sample) with stage IV colorectal carcinoma. In total, we were able to isolate 37 intact CTCs from six patients and identied in those multiple colorectal cancerassociated copy number changes, many of which were also present in the respective primary tumor. We then used massive parallel sequencing of a panel of 68 colorectal cancerassociated genes to compare the mutation spectrum in the primary tumors, metastases, and the corresponding CTCs from two of these patients. Mutations in known driver genes [e.g., adenomatous polyposis coli (APC), KRAS, or PIK3CA] found in the primary tumor and metastasis were also detected in corresponding CTCs. However, we also observed mutations exclusively in CTCs. To address whether these mutations were derived from a small subclone in the primary tumor or represented new variants of metastatic cells, we conducted additional deep sequencing of the primary tumor and metastasis and applied a customized statistical algorithm for analysis. We found that most mutations initially found only in CTCs were also present at subclonal level in the primary tumors and metastases from the same patient. This study paves the way to use CTCs as a liquid biopsy in patients with cancer, providing more effective options to monitor tumor genomes that are prone to change during progression, treatment, and relapse. Cancer Res; 73(10); 296575. Ó2013 AACR. Introduction Circulating tumor cells (CTC) are rare cells found in the blood of patients with solid tumors. The isolation and char- acterization of CTCs has tremendous potential for new bio- logic insight with very real clinical applications, such as the identication of prognostic, predictive, and pharmacokinetic biomarkers (1, 2). For example, in colorectal cancer, the KRAS mutations in exon 2 (codons 12 and 13) represent a paradigm that has been established as a negative predictive marker for treatment with EGF receptor (EGFR) inhibitors (3). Thus, "real- time" longitudinal monitoring of CTC-derived genotypes may provide a noninvasive approach to identify drug sensitivity and resistance-associated markers, guiding therapeutic decisions. However, because CTCs constitute as few as 1 cell per 1 10 9 normal blood cells in patients with metastatic cancer, it is difcult to identify and isolate these cells with current detec- tion methods (4). Multiple approaches to CTC isolation and characterization have been published (5, 6). However, the main application of the most currently available CTC detection systems consists of an enumeration of putative CTCs without further analyses. A powerful example of the application of CTC genotyping was provided by a study of patients with nonsmall cell lung cancer (NSCLC). A subset of NSCLC has somatic activating mutations in EGFR, and these patients are likely to benet from treatment with selective EGFR kinase inhibitors. Through monitoring of CTCs, the acquisition of the recurrent Authors' Afliations: 1 Institute of Human Genetics and 2 Institute of Pathology; Divisions of 3 Oncology and 4 Hematology, Medical University of Graz; 5 Department of Pathology, General Hospital Graz West, Graz, Austria; and 6 Institute of Tumor Biology, University Medical Center Ham- burg Eppendorf, Hamburg, Germany Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). E. Heitzer, M. Auer, C. Gasch, and M. Pichler contributed equally to this work. Corresponding Authors: Michael R. Speicher, Institute of Human Genet- ics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria. Phone: 43-316-380-4110; Fax: 43-316-380-9605; E-mail: [email protected]; and Jochen B. Geigl, E-mail: [email protected] doi: 10.1158/0008-5472.CAN-12-4140 Ó2013 American Association for Cancer Research. Cancer Research www.aacrjournals.org 2965 on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Transcript of Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from...

Page 1: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

Integrated Systems and Technologies

Complex Tumor Genomes Inferred from Single CirculatingTumor Cells by Array-CGH and Next-GenerationSequencing

Ellen Heitzer1, Martina Auer1, Christin Gasch6, Martin Pichler3, Peter Ulz1, Eva Maria Hoffmann1, Sigurd Lax5,Julie Waldispuehl-Geigl1, Oliver Mauermann6, Carolin Lackner2, Gerald H€ofler2, Florian Eisner3, Heinz Sill4,Hellmut Samonigg3, Klaus Pantel6, Sabine Riethdorf6, Thomas Bauernhofer3, Jochen B. Geigl1, andMichael R. Speicher1

AbstractCirculating tumor cells (CTC) released into blood from primary cancers and metastases reflect the current

status of tumor genotypes, which are prone to changes. Here, we conducted the first comprehensive genomicprofiling of CTCs using array–comparative genomic hybridization (CGH) and next-generation sequencing. Weused the U.S. Food and Drug Administration–cleared CellSearch system, which detected CTCs in 21 of37 patients (range, 1–202/7.5 mL sample) with stage IV colorectal carcinoma. In total, we were able to isolate37 intact CTCs from six patients and identified in those multiple colorectal cancer–associated copy numberchanges, many of which were also present in the respective primary tumor. We then used massive parallelsequencing of a panel of 68 colorectal cancer–associated genes to compare the mutation spectrum in theprimary tumors, metastases, and the corresponding CTCs from two of these patients. Mutations in knowndriver genes [e.g., adenomatous polyposis coli (APC), KRAS, or PIK3CA] found in the primary tumor andmetastasis were also detected in corresponding CTCs. However, we also observed mutations exclusively inCTCs. To address whether these mutations were derived from a small subclone in the primary tumor orrepresented new variants of metastatic cells, we conducted additional deep sequencing of the primary tumorand metastasis and applied a customized statistical algorithm for analysis. We found that most mutationsinitially found only in CTCs were also present at subclonal level in the primary tumors andmetastases from thesame patient. This study paves the way to use CTCs as a liquid biopsy in patients with cancer, providing moreeffective options to monitor tumor genomes that are prone to change during progression, treatment, andrelapse. Cancer Res; 73(10); 2965–75. �2013 AACR.

IntroductionCirculating tumor cells (CTC) are rare cells found in the

blood of patients with solid tumors. The isolation and char-acterization of CTCs has tremendous potential for new bio-logic insight with very real clinical applications, such as the

identification of prognostic, predictive, and pharmacokineticbiomarkers (1, 2). For example, in colorectal cancer, the KRASmutations in exon 2 (codons 12 and 13) represent a paradigmthat has been established as a negative predictive marker fortreatment with EGF receptor (EGFR) inhibitors (3). Thus, "real-time" longitudinal monitoring of CTC-derived genotypes mayprovide a noninvasive approach to identify drug sensitivity andresistance-associated markers, guiding therapeutic decisions.However, because CTCs constitute as few as 1 cell per 1 � 109

normal blood cells in patients with metastatic cancer, it isdifficult to identify and isolate these cells with current detec-tion methods (4). Multiple approaches to CTC isolation andcharacterization have been published (5, 6). However, themainapplication of the most currently available CTC detectionsystems consists of an enumeration of putative CTCs withoutfurther analyses.

A powerful example of the application of CTC genotypingwas provided by a study of patients with non–small cell lungcancer (NSCLC). A subset of NSCLC has somatic activatingmutations in EGFR, and these patients are likely to benefit fromtreatment with selective EGFR kinase inhibitors. Throughmonitoring of CTCs, the acquisition of the recurrent

Authors' Affiliations: 1Institute of Human Genetics and 2Institute ofPathology; Divisions of 3Oncology and 4Hematology, Medical Universityof Graz; 5Department of Pathology, General Hospital Graz West, Graz,Austria; and 6Institute of Tumor Biology, University Medical Center Ham-burg Eppendorf, Hamburg, Germany

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

E. Heitzer, M. Auer, C. Gasch, and M. Pichler contributed equally to thiswork.

Corresponding Authors: Michael R. Speicher, Institute of Human Genet-ics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria.Phone: 43-316-380-4110; Fax: 43-316-380-9605; E-mail:[email protected]; and Jochen B. Geigl, E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-12-4140

�2013 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 2965

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 2: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

T790M-EGFR drug resistance mutation has become evident inCTCs during the course of therapy and has coincided with thedevelopment of clinically refractory disease (7).

In fact, single-cell analyses have evolved to an active area,with various attempts to assess the genome of individual cells.For example, single-nucleus sequencing was shown to becapable of characterizing tumor evolution in breast cancer(8). However, this method was not suitable for assessing thegenetic characteristics of single tumor cells at a single-nucle-otide resolution because sequencing was conducted at merelyapproximately 0.2 times coverage. Therefore, only copy num-ber variations in single tumor cells were analyzed (8). In otherstudies, individual cells fromhematopoietic (9) and kidney (10)tumors were analyzed by exome amplification and subsequentsequencing. Obtaining a 30 times coverage, the data could beused to establish lineage relationships between the cells, basedon the point mutations (9, 10). These studies also showedthat amplification and sequencing errors are a concern forsingle-cell mutation analysis, as the false discovery ratio was2–3 � 10�5 and the average allele dropout ratio for all singlecells was approximately 11% (9, 10). To limit these problems,the authors defined a true mutation only if it occurred in aspecified number of cancer cells. For example, somatic-mutantloci had to be present in at least 5 myeloproliferative tumorcells and be homozygous normal in the respective nonmalig-nant tissue used for comparison (9). More recently, a single-cellwhole-genome analysis to measure the genomic diversity inone individual's gamete genome was described to createpersonal recombinationmaps fromwhole-genome sperm data(11). Using a special microfluidic device on haploid cells, theauthors reduced the false discovery rate to 4 � 10�9 with 5times coverage.

These studies suggest that high-resolution single-cell anal-ysis is feasible. However, compared with the aforementionedtechnologies, single-cell CTC analyses differ in 4 importantaspects. First, somatic mutations in tumor cells include basesubstitutions, indels, structural rearrangements, and changesin the copy number of DNA segments (12). However, no single-cell approach that effectively combines the reliable mapping ofcopy number changes with next-generation sequencingapproaches to detect nucleotide changes has been publishedso far. Second, CTCs are extremely rare events and, despitemany efforts (2, 13, 14), there is still a lack of technologiescapable of isolating them in sufficient numbers. As a conse-quence, usually only very limited numbers of cells are availablefor analysis. This excludes the aforementioned strategies, suchas calling mutations only if they are observed in a specifiednumber of cells. Third, isolation of CTCs out of millions ofnormal cells is a much more complicated procedure thanisolation of cells from a primary tumor or a selection of spermcells. Fourth, no suitable material for comparison of CTCresults is available. This is because CTCs may recur years afterinitial diagnosis of the primary tumor and may have acquiredmultiple novel changes since then. Furthermore, CTCs may bereleased from various metastatic sites, and their origin canusually not be traced. In addition, CTCs have been reported tobe heterogeneous (15–17) and may therefore exhibit a tremen-dous cell-to-cell variability.

For these reasons, the detailed characterization of CTCs isstill in its infancy. To date, only a few examples exist in whichhigh-dimensional analyses of individual CTCswere conducted,and these were analyses of CTC gene expression (18, 19). Here,we carried out a pilot study to investigate strategies for copynumber analysis and next-generation sequencing of singleCTCs. Our results suggest that complex tumor genomes canbe reconstructed from the peripheral blood of patients withcancer, and this approach may pave the way for new diseasemonitoring strategies.

Materials and MethodsPatients

All patients had advanced-stage [Union Internationale Con-tre le Cancer (French) stage IV] progressive disease at the timeof blood collection. Clinical characteristics are summarized inSupplementary Table S1. The study was approved by the localethics committee, and written informed consent was obtainedfrom all the patients. The patients were seen at the Division ofClinical Oncology, Department of Internal Medicine, MedicalUniversity of Graz (Graz, Austria).

Detection of CTCs and whole-genome amplificationBlood samples (7.5-mL each) were collected into CellSave

tubes (Veridex, Raritan). The Epithelial Cell Kit (Veridex) wasapplied for CTC enrichment and enumeration with the Cell-Search system as described previously (20). In brief, in a firststep, CTCs were captured by antiepithelial cell adhesionmolecule (EpCAM) antibody-bearing ferrofluid. Subsequentidentification of CTCs was based on cytokeratin positivity andnegativity for the leukocyte common antigen CD45. In addition,40, 6-diamidino-2-phenylindole staining was done to evaluatethe integrity of the nucleus. The further processing of CTCs isdescribed in the Supplementary Materials and Methods.

Enrichment of genes and 454 Life Sciences/RocheDiagnostics

The enrichment of genes and subsequent sequencing isoutlined in the Supplementary Materials and Methods.

Deep sequencing with the Illumina MiSeqFor validation of "private CTC mutations", i.e., mutations

found in only one CTC, we employed a comparative ultradeepsequencing approach using the Illumina MiSeq as exemplifiedin the Supplementary Materials and Methods.

ResultsPatients

In this study, we used the U.S. Food andDrug Administration(FDA)–approvedCellSearch system for CTCdetection (20) in 37patients with stage IV colorectal cancer. In 5 (13.5%) cases,analysis was not possible due to technical reasons, for exampleclotting of the blood. In 11 (29.7%) cases, noCTCswere found, in15 (40.5%) cases between 1 and 10 CTCs were found, and in 6(16.2%) cases more than 10 CTCs were identified. In the latter 6cases (i.e., patient #6, #9, #18, #22, #26, and #38), we isolatedCTCs for further analyses. For these 6 patients, the intervalbetween diagnosis of the primary tumor and blood collection to

Heitzer et al.

Cancer Res; 73(10) May 15, 2013 Cancer Research2966

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 3: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

obtain CTCs varied, with a range from 1 month (patient #9 and#18) to 3 years and 7 months (45 months, patient #38; mean, 15months; median, 5 months). In addition, clinical characteristicsof these patients are summarized in Supplementary Table S1.

Establishment of copy number changes in CTCsIn a next step, wewanted to test whether tumor-specific copy

number changes are reflected in CTCs. To this end, we used ourpreviously published whole-genome amplification (WGA) andarray–comparative genomic hybridization (CGH) technologiesfor analysis of a single cell or a few cells (21–27). Altogether, weanalyzed 37 CTCs and, as a control, we isolated leukocytes(CD45-positive cells). As expected, the CD45-positive cellsshowed balanced array-CGH profiles (data not shown).Our analysis consisted of several steps, which we carried

out for each CTC of each patient. We use patient #26 as an

example to explain these steps. The primary tumor ofpatient #26 had initially been diagnosed and completelyremoved 34 months before our analysis. A liver metastasiswas noted and resected 10 months after the initial diagnosis.Twenty-one months later, metastases were identified in theliver, spleen, and bone. We started our analyses by compar-ing the ratio of the profiles of the available tumor material, inthis case, the primary tumor (Fig. 1A), with the liver metas-tasis (Fig. 1B). We observed several common copy numberchanges, including, for example, losses of chromosomes 4,5q13.2–5q31.2, which harbors the adenomatous polyposiscoli (APC) gene, 8p, 17, and 18, and gains of chromosomes 8q,9, and 20. However, some differences were also observed,such as losses on chromosomes 10, 11, and 12 or gain ofchromosome 7, which were only present in the metastasisbut not in the primary tumor.

Figure 1. Analysis of the primary tumor, metastasis, and one circulating tumor cell (CTC05) from patient #26. A, ratio profile of the primary colorectal cancertumor of patient #26. The single green and red bars summarize the regions that were gained or lost based on all iterative calculations of our algorithm(Supplementary Information). The black profile regions represent balanced regions, lost regions appear in red, andgained regions are shown ingreen.B, array-CGH of the liver metastasis of patient #26. C, ratio profile of one representative CTC, i.e., CTC05. D, heatmaps comparing the copy number changes in theprimary tumor (PT), metastasis (Met.), and CTC05 (black, balanced regions; red, underrepresented regions; green, overrepresented regions). E, the bar chartdisplays the percentages of chromosomal regions that were commonly lost (red), balanced (black), or gained (green) in all 3 samples, i.e., primary tumor,metastasis, and CTC05, shared by metastasis and CTC05 only (blue), shared by primary tumor and CTC05 only (yellow), or unique to CTC05 (gray).

CTC Analysis by Array-CGH and Next-Generation Sequencing

www.aacrjournals.org Cancer Res; 73(10) May 15, 2013 2967

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 4: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

Accordingly, CTCs showed similar copy number changescompared with both primary tumor and metastasis but alsosome others, such as gains of chromosome 10 and 13 inCTC05 (Fig. 1C). For our array-CGH, we used a 60-Kmicroarray platform consisting of 59,012 oligonucleotidesfor analysis, and we calculated for each of these oligonu-cleotides whether the ratio values were decreased, bal-anced, or increased and illustrated the results in heatmaps(Fig. 1D). Finally, we determined whether the copy numberstatus for each oligonucleotide occurred only in the respec-tive CTC or also in the primary tumor and/or metastasis(Fig. 1E). This detailed analysis revealed that the copynumber status of 60.5% of the oligonucleotides on our

array platform was identical in the primary tumor andmetastasis as well as in CTC05 (i.e., of all 59,012 oligonu-cleotides on our array, 8.7% oligonucleotides showed a loss,42.9% remained balanced, and 8.9% showed a gain in copynumber status); 9.3% of the oligonucleotides were sharedonly by the primary tumor and CTC05, 12.6% were sharedonly by the metastasis and CTC05, and 17.6% were uniqueto CTC05 (Fig. 1E). These findings suggested that theobserved changes in the CTC05 were tumor specific andconsisted of changes also present in the primary tumor andthe liver metastasis, and in addition some other changes.

To investigate the CTC population structure, we extendedthese analyses to 10 CTCs from patient #26 (Fig. 2A).

Figure 2. Analysis of 10 CTCs from patient #26. A, percentages of oligonucleotides that were commonly lost (red), balanced (black), or gained (green) in theprimary tumor, metastasis, and respective CTC. Identical copy number changes occurring only in metastasis and respective CTCs are shown in blue andidentical changes in both primary tumor and CTC are shown in yellow. Copy number changes, which were observed only in the CTCs but not inthe primary tumor or metastasis, are displayed in gray. On average, for these 10 CTCs, 52.0% (median, 53.4%; range, 37.3%–61.5%) of the copy numberstatus changes (i.e., gains, losses, and balanced regions) were present in all CTCs, primary tumors, and analyzed metastases; 8.3% (median, 8.2%; range,6.7%–10.4%) were partially shared between all CTCs and the metastasis only; 14.2% (median, 14.2%; range, 12.0%–16.3%) were partially shared betweenthe CTCs and primary tumor only; and 25.6% (median, 24.8%; range, 17.6%–41.9%) of all CTC copy number changes were not observed in the primarytumor or metastasis. B, integer copy number profile of CTC26. C, combined copy number profile representing an average profile from all individualCTCs. D, hierarchical cluster analysis to determine the CTC population substructure and the relationship of CTCs to the primary tumor and metastases.mCTCl, main CTC lineage, i.e., the average copy number profile from all CTCs (black, balanced regions; red, underrepresented regions; and green,overrepresented regions).

Heitzer et al.

Cancer Res; 73(10) May 15, 2013 Cancer Research2968

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 5: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

Furthermore, assuming that single cells would have distinctcopy number states, we calculated integer copy numberprofiles (Supplementary Methods) for each CTC. This anal-ysis revealed that all 10 CTCs were within the tetraploidrange (e.g., CTC26, Fig. 2B; additional CTCs are depicted inSupplementary Fig. S1A–S1D). To establish the most fre-quently occurring copy number changes within the CTCpopulation of patient #26, we constructed an average copynumber profile from all individual CTCs (SupplementaryMethods; Fig. 2C). The "main CTC lineage" differed from theprimary tumor and its metastasis (Fig. 1A and B) by gains onchromosomes 1q, 6q, and 12p, a high-level gain on chromo-some 13, and loss on chromosome 14. When we conductedhierarchical clustering from the copy number profiles (Fig.2D), the integer copy number profile of one cell (i.e., CTC28;Supplementary Fig. S1D) deviated from that of the othercells, suggesting the presence of cells with a "private" patternof alterations.

CTCs display multiple colorectal cancer-associated copynumber changes

Weextended these analyses toCTCs from the other patients.In patient #6, we noted extensive differences between the copynumber profiles of the primary tumor and its metastasis(Supplementary Fig. S2A and S2B; details to patient's #6 historyare in Supplementary Information). From patient #6, weanalyzed 9 cells. In 3 cells, we did not find the KRAS G12Vmutation previously identified in the primary tumor, and inthese cells we did not observe any tumor-related copy numberchanges. From the 6 other cells that contained the KRASmutation, 2 cells were in the diploid range with few copynumber changes, whereas 4 cells were tetraploid and hadmultiple copy number changes (Fig. 3A). Interestingly, theaverage CTC copy number profile (Fig. 3B) differed from theprimary tumor (Supplementary Fig. S2A) by a gain of chro-mosome 8q, a numerical change that has frequently beendescribed in colorectal cancer (www.progenetix.net). Our

Figure 3. Detailed analysis of the 4 CTCs from patient #6. A, integer copy number profile of CTC07. B, combined copy number profile representing anaverage copy number profile from the 4 tetraploid CTCs from patient #6. C, percentages of oligonucleotides with copy number changes in comparisonwith the primary tumor, metastasis, and respectiveCTC. Identical copy number changes occurring only inmetastasis and respectiveCTCs are shown in blue,and identical changes in both primary tumor and CTC are shown in yellow. Copy number changes that were observed only in the CTCs but not in theprimary tumor or metastasis are displayed in gray. D, hierarchical cluster analysis (black, balanced regions; red, underrepresented regions; green, over-represented regions).

CTC Analysis by Array-CGH and Next-Generation Sequencing

www.aacrjournals.org Cancer Res; 73(10) May 15, 2013 2969

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 6: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

analyses revealed that the CTCs had more chromosomal copynumber changes in commonwith the primary tumor thanwiththe cerebellar metastasis (Fig. 3C). Again, cluster analysis (Fig.3D) identified a cell with a "private" copy number profile (i.e.,CTC14; Supplementary Fig. S2C).

At the time of diagnosis, patient #9 already had metastasesin liver, bone, and abdomen (peritoneal carcinomatosis).We obtained biopsies from the primary tumor (SupplementaryFig. S3A) and the peritoneal carcinomatosis (SupplementaryFig. S3B), which revealed almost identical array-CGH profilesfrom both sites. We conducted our CTC analyses 1 monthafter diagnosis and observed many similarities between thecopy number profiles of the CTCs and those from theprimary tumor and peritoneal carcinomatosis, respectively(Fig. 4).

We analyzed 6 epithelial and CD45-negative cells selected bythe CellSearch system from patient #38 (Supplementary Fig.S4A and S4B; history in Supplementary Information). However,we were unable to identify the KRASG12Dmutation, identified

in the primary tumor, in any of these cells. Furthermore, all ofthese cells had a balanced profile.

In 2 further cases (#18, #22) only small biopsies were taken atthe time of diagnosis, so that insufficientmaterial was availableto analyze the primary tumor for comparison. However, intheir CTCs, we again identified multiple copy number changesassociated with colorectal cancer (as documented at www.progenetix.net). For example, in patient #22, one CTC revealedlosses on chromosomes 3, 4, 5, 8p, and 18 and gains onchromosomes 7p, 17q, and 20 (Supplementary Fig. S5; CTC03).

Sequencing of the ultraconserved region UCR41 in CTCsTo establish appropriate CTC sequencing strategies, we

selected CTCs from patients #6 and #26 because we had thelargest CTC number for these 2 patients. However, sequencingof single-cell amplification products can be prone to experi-mental and technical errors (9–11). To address this issue, weexploited ultraconserved regions (UCR), which represent geno-mic regions that do not accumulate true mutations (28). Thus,

Figure 4. Summary of CTC analysis of patient #9. A, integer copy number profile of CTC 4. B, combined copy number profile representing an average copynumber profile frompatient #90s four CTCs. C, percentages of oligonucleotideswith copy number changes in comparisonwith the primary tumor,metastasis,and respective CTC. D, hierarchical cluster analysis.

Heitzer et al.

Cancer Res; 73(10) May 15, 2013 Cancer Research2970

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 7: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

all sequence variations in UCRs should represent bona fideerrors, making UCRs an ideal control for DNA amplificationand sequencing errors (28). Therefore, we sequenced theultraconserved region UCR41 (ref. 29; Supplementary Fig. S6,green bar), its adjacent extremely conserved region (Supple-mentary. Fig. S6, gray bar), and nonconserved flanking regions(28) and analyzed 5 cells each from healthy male and femalecontrols. The only sequence variant in single cells from healthycontrols was at single-nucleotide polymorphism (SNP)rs17701179, where the male donor had A/G alleles, which wealso identified in each of his 5 single cells (Supplementary Fig.S6, blue arrows).Next, we analyzed the constitutional DNA, primary tumor,

metastasis, and all the CTCs from patients #6 (n¼ 4) and #26(n ¼ 10), using the same procedure. Again, we found nosequence variation in the ultraconserved or extremely con-served regions. However, we identified 2 variations in thenonconserved flanking regions. One was in the metastasissample from patient #6 (Supplementary Fig. S6, right redarrow), in which the sample had not been subjected to WGAbut only to the amplification steps necessary for the targetedgene enrichment. The second variation was in one of the 10CTCs from patient #26 (CTC21; Supplementary Fig. S6, leftred arrow). Both of these variants were within a region thatseems to have increased mutability compared with the aver-age mutability (28). Furthermore, patient #6 had constitu-tional A/G alleles for SNP rs3910657; however, we identifiedA/A in CTC7 and G/G in CTC14. Our results suggest that,despite the amplification steps involved in our protocol, nosequencing artifacts occurred in this ultraconserved region.

Mutation spectrum in primary tumors, metastases, andCTCsWe then aimed at establishing a mutation spectrum for a

panel of 68 colorectal cancer-related genes (SupplementaryTable S2) that are frequently (> 3%) mutated in colorectalcancer according to the COSMIC database. We analyzed these68 genes in the different samples, including constitutional DNA,the primary tumors, and theirmetastases in patients #6 and#26.In addition, we sequenced 3CTCs (7, 13, and 14) frompatient #6and 5 CTCs (5, 21, 22, 24, and 28) from patient #26.A total of 959,790 and 2,070,381 sequences were generated

for the formalin-fixed paraffin-embedded (FFPE) pool (tumorand metastasis of patient #6 and #26) and the 2 CTC pools(pool 1: CTC 7, 13, 14 from patient #6 and CTC 5 from patient#26; pool 2: CTC 21, 22, 24, 28 from patient #26), respectively.Read length was remarkably consistent throughout the exper-iment. Theaverage read lengthwas 232 bp for the FFPEmaterialand 329 bp for the CTCs, and the total yield from the sequencingrun was 223 Mb for the 4 FFPE samples and 683 Mb for the 8CTCs. The average coverage of tumors and metastases (�24)was lower as compared with the CTCs (�41). Sequencing ofenriched CTCs achieved more on-target reads (55.6%) than didsequencing of the FFPE material (41.5%).With the exception of an exonic deletion in ERCC6L in

patient #6 and a 1-bp insertion in MLH1 in patient #26 (inCTC28), all identified insertions and deletions were found onlyin introns.We focused our analysis on exonic, nonsynonymous

variants not included in the National Center for BiotechnologyInformation dbSNP build 132 database. In the initial screen, wefound 25 mutations in 17 genes in at least one of the analyzedsamples of patients #6 and 22 mutations in 10 genes in patient#26. Sanger sequencing confirmed 16 and 15 mutations inpatients #6 (Fig. 5A) and #26 (Fig. 5B), respectively (Supple-mentary Table S3). In patient #6, 4 of these mutations wereconstitutional and therefore found in all the samples. Thispatient also had 3 somatic mutations in known colorectalcancer driver genes (i.e., in APC, KRAS, and PIK3CA), whichwere present in all the tumor samples including CTCs. Inter-estingly, we found 2 additional mutations in the cerebellarmetastasis in NF1 and TP53, which we also detected in the 3CTCs. The other mutations found were present only in singleCTCs examined (Fig. 5A).

In patient #26, we identified one constitutional mutation inall analyzed samples. However, within our panel of genes, wedid not find any othermutation present in all samples (Fig. 5B).However, array-CGH had identified a deletion on chromosome5 harboring theAPC gene in all tumor samples, and therefore inthis case copy number changes may indicate genomic regions,which had been involved in tumor initiation and/or progres-sion. In patient #26, we again identified "private mutations,"defined as mutations observed in only one CTC but not in anyother tumor material. Because of the high experimental errorrate of single-cell analysis, we carried out additional experi-ments to address the nature of these mutations as outlinedbelow.

Most private CTC mutations are present at a subclonallevel in the primary tumor

We reasoned thatmutations found in only one CTC could beamplification/sequencing artifacts or mutations already pres-ent in the primary tumor or metastasis at subclonal level andtherefore they could have been missed by our initial sequenc-ing efforts. To address this question, we conducted ultradeepsequencing of the primary tumors, metastases, and normaltissues to test whether private CTC mutations might reflectsubclonal mutations. Subclonal single-nucleotide variantswere detected and quantified with the deep single-nucleo-tide variant (SNV) algorithm, an approach capable of detect-ing variants with frequencies as low as one per 10,000 alleles(30). These analyses revealed that from 20 analyzable privateCTC mutations (one variant was not amplifiable), 17 (85%)were present in at least one other corresponding sample (i.e.,primary tumor or metastasis) with mutation frequenciesranging from 0.02 to 0.42 (Fig. 5; Table 1). For the remainingmutations, it is impossible to decide whether these are truemutations occurring with allele frequencies below the res-olution limits of current detection methods or if they areamplification or sequencing artifacts.

Correlation with clinical parameters and potentialclinical applications

Although this was a pilot study, we then attempted toinvestigate whether the identified set of genetic aberrationscould potentially be informative for identifying rationaltherapies currently available or in clinical trials. Mutations

CTC Analysis by Array-CGH and Next-Generation Sequencing

www.aacrjournals.org Cancer Res; 73(10) May 15, 2013 2971

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 8: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

of biologic or predictive relevance are listed in Fig. 5A and B.For example, 2 mutations in patient #6 are of clinicalrelevance, i.e., KRAS p.G12V (resistant to EGFR monoclonalantibodies) and PIK3CA p.E542K [an exon 9 activatingmutation; possible predictor for EGFR antibodies and/orinsulin-like growth factor (IGF)-1R inhibitors; ref. 31].Although the CTC analysis confirmed that these mutationswere still present 8 months after diagnosis, it did not revealnovel targets for therapeutic interventions as compared withthe analysis of the primary tumor. In contrast, in patient #26,we identified cyclin-dependent kinase 8 (CDK8) amplifica-tion on chromosome 13q12.13 in 9 of 10 CTCs that was notpresent in the parts of the primary tumor or metastasis thatwe analyzed. This amplification may represent a viabletarget for CDK inhibitors, which are currently in clinicaltrials (32–34).

DiscussionA major goal of cancer medicine is to move from fixed

treatment regimens to therapies tailored to a patient'sindividual tumor. In this study, we addressed whethercomplex tumor genomes can be inferred noninvasively fromCTCs of patients with cancer. Detailed characterization ofCTCs may provide the opportunity for clinical impact inthe treatment of selected cancers in which appropriatetargeting of tumor-associated genetic lesions is critical.Such an approach would allow monitoring of tumor gen-omes with an unprecedented resolution. However, thescience of CTCs is still in its infancy. Little is known aboutthe biology of these cells, and only recently technologicadvances have provided a window into the composition ofthis rare cell population, allowing molecularly designedanalyses (18, 19).

Figure 5. Summary of mutations identified in patients #6 (A) and #26 (B) in the respective analyzed material. Somatic mutations found in all tumor samples areindicated in red, mutations are shown in blue if they were present in all samples except the primary tumor, and the other mutations are shown inblack. The constitutional mutation, which we identified in patient #26, is shown in green. Mutations occurring in only one CTC, for which deep sequencing didnot provide evidence for existence at a subclonal level in the primary tumor or metastasis, are shown in gray. The tables highlight mutations or copy numberchanges of particular biologic or clinical interest. The column "Copy number CTCs" refers to the average copy number of all analyzed CTCs.

Heitzer et al.

Cancer Res; 73(10) May 15, 2013 Cancer Research2972

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 9: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

The importance of a molecular characterization of thesecells is underlined by the fact that we had 9 epithelial andCD45-negative cells selected by the CellSearch system (3 frompatient #6 and 6 from patient #9), in which we did not find theKRASmutation previously identified in the respective primarytumors and which each had a balanced copy numberprofile. Formally, we cannot exclude that these cells are CTCs;however, alternatively these cells could be circulating epithelialcells, which have recently been described in patients withbenign colon diseases (35). Thus, a simple enumeration ofepithelial, CD45-negative cells without molecular characteri-zation for prognostic purposes may in some cases beerroneous.Although the CellSearch system used for CTC enrichment

has been cleared by the FDA, it has shortcomings, as theenrichment is based on EpCAM expression of CTCs andEpCAM-negative CTCs are missed. Increasing evidence sug-gests that EpCAM-negative CTCs might have undergone anepithelial-to-mesenchymal transition, a process linked to the

stemness of cancer cells and increased chemoresistance (36).Other emerging CTC isolation techniques, including non-EpCAM or size-based enrichment approaches (5), have theadvantage of assessing the relevant genomic changes in abroader range of CTC subsets. However, these new CTC assayshave not been validated to a comparable level as the CellSearchsystem with regard to their specificity, reproducibility, andclinical relevance so far. Moreover, recent reports indicate thatcarcinoma cells have to express epithelial markers (such asEpCAM) to colonize distant organs and to form overt metas-tases (36).

We applied our single-cell technologies (21–27) to a detailedanalysis of CTCs and showed for the first time that mutationanalysis of multiple genes by next-generation sequencing incombination with establishing the copy number status isfeasible from CTCs. Our strategy differs from other recentlypublished single-cell approaches as these have shortcomingsfor the analysis of rare cell events. For example, single-cellsequencing (8) allows only assessment of copynumber changes

Table 1. Results of ultradeep sequencing with DNA from primary tumor andmetastases of patients #6 and#26, respectively

Primary tumor Metastasis

GeneAmino acidchangea

Sampleb andpatient % Mutatedc Total reads % Mutatedc Total reads

ABCA1 p.P1209S CTC7,#6 0.05 950,656 0.08 480,081C10orf137 p.A990E CTC14,#6 0.02 938,780 not significantd

CACNA2D3 p.G84S CTC14,#6 0.07 842,856 0.07 996,436CTNNB1 p.A149T CTC14,#6 0.08 547,498 0.42 432,851GNAS p.G869D CTC14,#6 not significantd 0.10 109,096GUCY1A2 p.H439Y CTC14,#6 0.06 299,872 0.02 299,872LAMA1 p.P1414S CTC7,#6 0.16 433,621 0.11 607,163LAMA1 p.H1002Y CTC7,#6 not significantd not significantd

ABCA1 p.A1714T CTC21,#26 0.04 315,684 0.11 153,447ADAMTSL3 p.Q756X CTC24,#26 0.20 258,482 0.02 242,142CTNNB1 p.L160I CTC5,#26 0.07 546,086 0.06 433,461CTNNB1 p.G277D CTC5,#26 0.18 374,324 0.03 118,076CTNNB1 p.C429Y CTC24,#26 0.10 155,213 0.05 108,917CSMD3 p.V1723I CTC22,#26 not significantd 0.21 247,608GNAS p.G869D CTC5,#26 not significantd not significantd

KIAA1409 p.S1344P CTC21,#26 not significantd not significantd

LAMA1 p.W818X CTC5,#26 not significantd 0.02 260,256MLH1 p.R497Pfs�6 CTC28,#26 not significantd not significantd

NAV3 p.G1921S CTC21,#26 0.08 359,151 0.03 190,238NAV3 p.S594F CTC5,#26 0.08 125,637 not significantd

OR51E1 p.A312V CTC21,#26 not analyzablee not analyzablee

NOTE: The columns "Gene" and "Amino acid change" list all private CTC mutations, i.e., mutations found in only one CTC. Thepercentages ofmutatedDNA fragments (%Mutated) and the total number of reads (Total reads) are given for the respective samples ofpatients #6 and #26, respectively.aHuman Genome Variation Society mutation nomenclature.bCTCs of patient 6 (#6) or patient 26 (#26).c% Mutated, percentage of total number of mutated reads (Phred quality of >15) calculated with the deepSNV algorithm.dNot significant,P value fromdeepSNValgorithm for the likelihoodcalculationwhether anobservedvariant is true or a sequencing errorwas not significant after Benjamini–Hochberg correction.eNot analyzable, PCR amplification failed.

CTC Analysis by Array-CGH and Next-Generation Sequencing

www.aacrjournals.org Cancer Res; 73(10) May 15, 2013 2973

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 10: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

but not of changes at the nucleotide level. The single-cellexome sequencing methods (9, 10) defined true mutationsonly if they occurred in a specified number of cancer cells. Thisis applicable for the analysis of cells from a primary tumor, inwhich a virtually unlimited number of cells is available foranalysis, but not for rare events, such as CTCs, in whichfrequently less than 5 cells with sufficient quality are available.Furthermore, this approach does not allow a genome-widemeasurement of copy number changes. The single-cell whole-genome analysis presented byWang and colleagues (11) seemsto be especially attractive as it may significantly reduce thefalse-discovery rate. However, it is currently unknown whetherthe microfluidic device used by these authors is applicable tothe identification and isolation of CTCs. Very recently, a newamplification method offering high uniformity across thegenome and also allowing both the genome-wide detectionof single-nucleotide and copy-number variations of a singlehuman cell has been reported (37, 38). Although this newamplification method has not been applied to CTCs yet, itseems to have great potential to advance CTC research further.

In any case, the aforementioned studies have documentedthat single-cell analysis is prone to artifacts, which may beintroduced either during amplification or sequencing (9–11, 37, 38). To address this issue, wemade use of a comparativesequencing strategy. This strategy involved reanalysis of theprimary tumor and metastasis by deep sequencing. The samegenomic region was compared between a heterogeneous testsample (i.e., tumor and metastasis) and a homogeneous con-trol sample (i.e., normal tissue). Sequencing results wereevaluated with deepSNV (30). With these sequencing effortsand statistical evaluations, variants occurring with allele fre-quencies ranging from 0.0002 to 0.34 can be identified in aheterogeneous tissue (30). Interestingly, these analyses sug-gested that the majority of private CTC mutations had alreadybeen present at a subclonal level in the primary tumor.Therefore the majority of these variants were likely truemutations and not artifacts.

Our study suggests that we can elucidate relevant changes inthe tumor genome that had either not been present or not beenobserved at the time of initial diagnosis. Thus, an importantapplication may be the detailed reevaluation of patientsenrolled in clinical trials for novel substances. For example,analysis of the genomes of the primary tumor andmetastasis ofpatient #26 did not reveal changes that would have made hereligible for enrollment in a trial with a CDK inhibitor such asflavopiridol (32–34). Yet, the CTCs obtained 34 and 24 monthsafter diagnosis of the primary tumor and liver metastasis,respectively, revealed a high level of amplification of CDK8,which had not been noted in the previous analyses. CDK8 hasbeen described as a positive regulator of catenin signalingin 15% to 20% of colorectal cancers (39). Furthermore, CDK8inhibition suppresses damage-induced tumor-promoting

paracrine activities of tumor cells, and it has recently beenreported that CDK8 inhibitors offer a promising approach toincrease the efficacy of cancer chemotherapy (40). Thereforethis amplification may represent a viable target for CDKinhibitors, which are currently in clinical trials (32–34).Although we cannot yet provide treatment outcomes, patient#26 may serve as an example of the impact of reevaluation ofthe tumor genome status on treatment decisions.

The next challenge will be to test such approaches for theserial monitoring of patients with cancer and to evaluatetheir potential for the early-stage analysis of peripheralblood. However, our proof-of-concept study suggests thatminimal invasive access to tumor material immediatelybefore specific antitumor treatment initiation in advancedcases may become possible for reevaluation of the cancergenome and possibly tailored management of treatmentchoices.

Disclosure of Potential Conflicts of InterestH. Sill has commercial research grants fromGerot and Roche, and K. Pantel is

a consultant/advisory board member of Veridex. No potential conflicts ofinterest were disclosed by the other authors.

Authors' ContributionsConception and design: E. Heitzer, M. Pichler, K. Pantel, T. Bauernhofer, J.B.Geigl, M.R. SpeicherDevelopment of methodology: E. Heitzer, M. Auer, C. Gasch, J. Waldispuehl-Geigl, O. Mauermann, K. Pantel, M.R. SpeicherAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M. Pichler, S. Lax, J. Waldispuehl-Geigl, C. Lackner,Gerald H€ofler, F. Eisner, H. Samonigg, K. Pantel, S. Riethdorf, T. Bauernhofer, J.B.GeiglAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): E. Heitzer, M. Auer,M. Pichler, P. Ulz, S. Lax, H. Sill, K.Pantel, S. Riethdorf, M.R. SpeicherWriting, review, and/or revision of the manuscript: E. Heitzer, M. Auer, M.Pichler, S. Lax, Gerald H€ofler, H. Sill, K. Pantel, S. Riethdorf, T. Bauernhofer, J.B.Geigl, M.R. SpeicherAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): E.M. Hoffmann, F. Eisner, H. Samonigg,T. Bauernhofer, J.B. GeiglStudy supervision: E. Heitzer, M. Auer, H. Samonigg, K. Pantel, J.B. Geigl, M.R.Speicher

AcknowledgmentsThe authors thank Mag. Maria Langer-Winter for editing the article.

Grant SupportThis work was funded by the European Commission (GENINCA, contract no.

202230; to K. Pantel andM.R. Speicher), the Austrian Science Fund (FWF; grant#:P20338 and W 1226-B18, DKplus Metabolic and Cardiovascular Disease to M.R.Speicher), and by the COMET Center ONCOTYROL (M.R. Speicher). E.M.Hoffmann was supported by the Molecular Medicine PhD Program of theMedical University of Graz.

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 indicate thisfact.

Received November 7, 2012; revised January 15, 2013; accepted February 18,2013; published OnlineFirst March 7, 2013.

References1. Cristofanilli M, BuddGT, EllisMJ, StopeckA,Matera J,MillerMC, et al.

Circulating tumor cells, disease progression, and survival inmetastaticbreast cancer. N Engl J Med 2004;351:781–91.

2. Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al.Isolationof rare circulating tumour cells in cancer patients bymicrochiptechnology. Nature 2007;450:1235–9.

Heitzer et al.

Cancer Res; 73(10) May 15, 2013 Cancer Research2974

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 11: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

3. Walther A, Johnstone E, Swanton C, Midgley R, Tomlinson I, Kerr D.Genetic prognostic and predictive markers in colorectal cancer. NatRev Cancer 2009;9:489–99.

4. Pantel K, Brakenhoff RH, Brandt B. Detection, clinical relevance andspecific biological properties of disseminating tumour cells. Nat RevCancer 2008;8:329–40.

5. Alix-Panabieres C, Pantel K. Circulating tumor cells: liquid biopsy ofcancer. Clin Chem 2013;59:110–8.

6. Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumorcells: approaches to isolation and characterization. J Cell Biol 2011;192:373–82.

7. Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, ColluraCV, et al. Detection of mutations in EGFR in circulating lung-cancercells. N Engl J Med 2008;359:366–77.

8. Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J, et al.Tumour evolution inferred by single-cell sequencing. Nature 2011;472:90–4.

9. Hou Y, Song L, Zhu P, Zhang B, Tao Y, Xu X, et al. Single-cell exomesequencing and monoclonal evolution of a JAK2-negative myelopro-liferative neoplasm. Cell 2012;148:873–85.

10. Xu X, Hou Y, Yin X, Bao L, Tang A, Song L, et al. Single-cell exomesequencing reveals single-nucleotide mutation characteristics of akidney tumor. Cell 2012;148:886–95.

11. Wang J, Fan HC, Behr B, Quake SR. Genome-wide single-cell analysisof recombination activity and de novomutation rates in human sperm.Cell 2012;150:402–12.

12. Stratton MR. Exploring the genomes of cancer cells: progress andpromise. Science 2011;331:1553–8.

13. Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, et al.Isolation of circulating tumor cells using a microvortex-generatingherringbone-chip. Proc Natl Acad Sci U S A 2010;107:18392–7.

14. Issadore D, Chung J, Shao H, Liong M, Ghazani AA, Castro CM, et al.Ultrasensitive clinical enumeration of rare cells ex vivo using a micro-hall detector. Sci Transl Med 2012;4:141ra92.

15. Gangnus R, Langer S, Breit E, Pantel K, Speicher MR. Genomicprofiling of viable and proliferative micrometastatic cells from early-stage breast cancer patients. Clin Cancer Res 2004;10:3457–64.

16. Kraus J, Pantel K, Pinkel D, Albertson DG, Speicher MR. High-reso-lution genomic profiling of occult micrometastatic tumor cells. GenesChromosomes Cancer 2003;36:159–66.

17. SolakogluO,MaierhoferC, LahrG,Breit E, ScheunemannP,Heumos I,et al. Heterogeneous proliferative potential of occult metastatic cells inbone marrow of patients with solid epithelial tumors. Proc Natl AcadSci U S A 2002;99:2246–51.

18. Powell AA, Talasaz AH, Zhang H, Coram MA, Reddy A, Deng G, et al.Single cell profiling of circulating tumor cells: transcriptional hetero-geneity and diversity from breast cancer cell lines. PLoS ONE 2012;7:e33788.

19. Stott SL, Lee RJ, Nagrath S, Yu M, Miyamoto DT, Ulkus L, et al.Isolation and characterization of circulating tumor cells from patientswith localized and metastatic prostate cancer. Sci Transl Med 2010;2:25ra3.

20. Riethdorf S, Fritsche H, Muller V, Rau T, Schindlbeck C, Rack B, et al.Detection of circulating tumor cells in peripheral blood of patients withmetastatic breast cancer: a validation study of the CellSearch system.Clin Cancer Re 2007;13:920–8.

21. Aleksic K, Lackner C, Geigl JB, Schwarz M, Auer M, Ulz P, et al.Evolution of genomic instability in diethylnitrosamine-induced hepa-tocarcinogenesis in mice. Hepatology 2011;53:895–904.

22. Begus-Nahrmann Y, Lechel A, Obenauf AC, Nalapareddy K, Peit E,Hoffmann E, et al. p53 deletion impairs clearance of chromosomal-

instable stem cells in aging telomere-dysfunctional mice. Nat Genet2009;41:1138–43.

23. Fiegler H, Geigl JB, Langer S, Rigler D, Porter K, Unger K, et al. Highresolution array-CGH analysis of single cells. Nucleic Acids Res 2007;35:e15.

24. Geigl JB, Obenauf AC, Waldispuehl-Geigl J, Hoffmann EM, Auer M,HormannM, et al. Identification of small gains and losses in single cellsafter whole genome amplification on tiling oligo arrays. Nucleic AcidsRes 2009;37:e105.

25. Geigl JB, Speicher MR. Single-cell isolation from cell suspensions andwhole genome amplification from single cells to provide templates forCGH analysis. Nat Protoc 2007;2:3173–84.

26. MathiesenRR, Fjelldal R, Liestol K, Due EU,Geigl JB, Riethdorf S, et al.High-resolution analyses of copy number changes in disseminatedtumor cells of patients with breast cancer. Int J Cancer 2012;131:E405–15.

27. Sperka T, Song Z, Morita Y, Nalapareddy K, Guachalla LM, Lechel A,et al. Puma and p21 represent cooperating checkpoints limitingself-renewal and chromosomal instability of somatic stem cells inresponse to telomere dysfunction. Nat Cell Biol 2011;14:73–9.

28. De Grassi A, Segala C, Iannelli F, Volorio S, Bertario L, Radice P, et al.Ultradeep sequencing of a human ultraconserved region revealssomatic and constitutional genomic instability. PLoS Biol 2010;8:e1000275.

29. Bejerano G, Pheasant M, Makunin I, Stephen S, Kent WJ, Mattick JS,et al. Ultraconserved elements in the human genome. Science 2004;304:1321–5.

30. GerstungM, Beisel C, RechsteinerM,Wild P, Schraml P,MochH, et al.Reliable detection of subclonal single-nucleotide variants in tumourcell populations. Nat Commun 2012;3:811.

31. De Roock W, De Vriendt V, Normanno N, Ciardiello F, Tejpar S. KRAS,BRAF, PIK3CA, and PTEN mutations: implications for targeted ther-apies in metastatic colorectal cancer. Lancet Oncol 2010;12:594–603.

32. Dickson MA, Shah MA, Rathkopf D, Tse A, Carvajal RD, Wu N, et al. Aphase I clinical trial of FOLFIRI in combination with the pan-cyclin-dependent kinase (CDK) inhibitor flavopiridol. Cancer ChemotherPharmacol 2010;66:1113–21.

33. Ramaswamy B, Phelps MA, Baiocchi R, Bekaii-Saab T, Ni W, Lai JP,et al. A dose-finding, pharmacokinetic and pharmacodynamic study ofa novel schedule of flavopiridol in patientswith advanced solid tumors.Invest New Drugs 2012;30:629–38.

34. Wang LM, Ren DM. Flavopiridol, the first cyclin-dependent kinaseinhibitor: recent advances in combination chemotherapy. Mini RevMed Chem 2010;10:1058–70.

35. Pantel K,DeneveE,NoccaD,Coffy A, Vendrell JP,MaudelondeT, et al.Circulating epithelial cells in patients with benign colon diseases. ClinChem 2012;58:936–40.

36. Joosse SA, Pantel K. Biologic challenges in the detection of circulatingtumor cells. Cancer Res 2013;73:8–11.

37. Lu S, ZongC, FanW, YangM, Li J, Chapman AR, et al. Probingmeioticrecombination and aneuploidy of single sperm cells by whole-genomesequencing. Science 2012;338:1627–30.

38. ZongC, Lu S, ChapmanAR, Xie XS. Genome-wide detection of single-nucleotide and copy-number variations of a single humancell. Science2012;338:1622–6.

39. Firestein R, Hahn WC. Revving the Throttle on an oncogene: CDK8takes the driver seat. Cancer Res 2009;69:7899–901.

40. Porter DC, Farmaki E, Altilia S, Schools GP, West DK, Chen M, et al.Cyclin-dependent kinase 8 mediates chemotherapy-induced tumor-promoting paracrine activities. Proc Natl Acad Sci U S A 2012;109:13799–804.

CTC Analysis by Array-CGH and Next-Generation Sequencing

www.aacrjournals.org Cancer Res; 73(10) May 15, 2013 2975

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140

Page 12: Complex Tumor Genomes Inferred from Single Circulating ... · Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing Ellen

2013;73:2965-2975. Published OnlineFirst March 7, 2013.Cancer Res   Ellen Heitzer, Martina Auer, Christin Gasch, et al.   Cells by Array-CGH and Next-Generation SequencingComplex Tumor Genomes Inferred from Single Circulating Tumor

  Updated version

  10.1158/0008-5472.CAN-12-4140doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2013/03/07/0008-5472.CAN-12-4140.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/73/10/2965.full#ref-list-1

This article cites 38 articles, 13 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/73/10/2965.full#related-urls

This article has been cited by 47 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://cancerres.aacrjournals.org/content/73/10/2965To request permission to re-use all or part of this article, use this link

on May 17, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 7, 2013; DOI: 10.1158/0008-5472.CAN-12-4140