Intratumor heterogeneity and homologous recombination ...

8
Intratumor heterogeneity and homologous recombination deciency of high-grade serous ovarian cancer are associated with prognosis and molecular subtype and change in treatment course Hisamitsu Takaya a , Hidekatsu Nakai a , Kazuko Sakai b , Kazuto Nishio b , Kosuke Murakami a , Masaki Mandai c , Noriomi Matsumura a, * a Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan b Department of Genome Biology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan c Department of Gynecology and Obstetrics, Kyoto University Graduateschool of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan highlights High intratumor heterogeneity in high-grade serous ovarian cancer associated with a shorter rst recurrent interval. High loss of heterozygosity score in high-grade serous ovarian cancer associated with a good prognosis. The number of the tumor clone decreased after chemotherapy and then increased at recurrence. Chemosensitivity are not always consistent with homologous recombination deciency. article info Article history: Received 17 August 2019 Received in revised form 2 November 2019 Accepted 8 November 2019 Available online 27 November 2019 Keywords: Ovarian cancer Clonality Loss of heterozygosity Chemo-resistance abstract Objective: High-grade serous ovarian cancers (HGSOC) are genomically characterized by homologous recombination deciency (HRD) and TP53 mutations, which lead to intratumor heterogeneity (ITH). This study aimed to reveal the relationship between HRD, ITH and prognosis and analyze their changes during treatment. Methods: We obtained 573 SNP array and gene expression array data from The Cancer Genome Atlas. SNP array data were processed to calculate the Clonality Index (CI) and loss of heterozygosity (LOH) scores. Gene expression array data were used for classifying molecular subtypes. Additionally, we ob- tained 33 samples from 20 HGSOC patients, including 4 samples from interval debulking surgery (IDS) and 9 samples from recurrent surgery. Results: We divided HGSOC samples into 2 groups. The high CI group showed a high recurrent risk, and the high LOH group showed a statistically good prognosis. Combining the two factors, the high LOH/low CI group showed a statistically good prognosis. In terms of molecular subtypes, the mesenchymal sub- type, which had a poor prognosis, showed a high CI with statisitically signicant difference and the immunoreactive subtype, which had a good prognosis, showed a tendency to have a high LOH score. Throughout treatment, the CI decreased to one at the IDS (n ¼ 4) and then increased at recurrence (n ¼ 3). LOH scores greatly decreased in two cases at the IDS. Conclusions: ITH and HRD were associated with prognosis in HGSOC. ITH decreased after neoadjuvant chemotherapy, suggesting that the chemo-resistant cancer clone remains after chemotherapy. © 2019 Elsevier Inc. All rights reserved. 1. Introduction Ovarian cancer has a poor prognosis among gynecologic ma- lignancies [1]. High-grade serous ovarian cancer (HGSOC), which is the most frequent epithelial ovarian cancer, is commonly diagnosed as an advanced cancer [2]. HGSOC is mainly treated by chemo- therapy; however, most HGSOCs recur as a chemo-resistant tumor Abbreviations: HGSOC, high-grade serous ovarian cancer; HRD, homologous recombination deciency; TCGA, The Cancer Genome Atlas; LOH, loss of hetero- zygosity; CI, clonality index; NAC, neoadjuvant chemotherapy. * Corresponding author. E-mail addresses: [email protected] (H. Takaya), [email protected] (H. Nakai), [email protected] (K. Sakai), [email protected] (K. Nishio), [email protected] (K. Murakami), [email protected]. ac.jp (M. Mandai), [email protected] (N. Matsumura). Contents lists available at ScienceDirect Gynecologic Oncology journal homepage: www.elsevier.com/locate/ygyno https://doi.org/10.1016/j.ygyno.2019.11.013 0090-8258/© 2019 Elsevier Inc. All rights reserved. Gynecologic Oncology 156 (2020) 415e422

Transcript of Intratumor heterogeneity and homologous recombination ...

Page 1: Intratumor heterogeneity and homologous recombination ...

Contents lists available at ScienceDirect

Gynecologic Oncology 156 (2020) 415e422

Gynecologic Oncology

journal homepage: www.elsevier .com/locate/ygyno

Intratumor heterogeneity and homologous recombination deficiencyof high-grade serous ovarian cancer are associated with prognosis andmolecular subtype and change in treatment course

Hisamitsu Takaya a, Hidekatsu Nakai a, Kazuko Sakai b, Kazuto Nishio b,Kosuke Murakami a, Masaki Mandai c, Noriomi Matsumura a, *

a Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japanb Department of Genome Biology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japanc Department of Gynecology and Obstetrics, Kyoto University Graduateschool of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan

h i g h l i g h t s

� High intratumor heterogeneity in high-grade serous ovarian cancer associated with a shorter first recurrent interval.� High loss of heterozygosity score in high-grade serous ovarian cancer associated with a good prognosis.� The number of the tumor clone decreased after chemotherapy and then increased at recurrence.� Chemosensitivity are not always consistent with homologous recombination deficiency.

a r t i c l e i n f o

Article history:Received 17 August 2019Received in revised form2 November 2019Accepted 8 November 2019Available online 27 November 2019

Keywords:Ovarian cancerClonalityLoss of heterozygosityChemo-resistance

Abbreviations: HGSOC, high-grade serous ovariarecombination deficiency; TCGA, The Cancer Genomzygosity; CI, clonality index; NAC, neoadjuvant chemo* Corresponding author.

E-mail addresses: [email protected] (H. Tak(H. Nakai), [email protected] (K. Sakai(K. Nishio), [email protected] (K. Murakac.jp (M. Mandai), [email protected] (N. Mats

https://doi.org/10.1016/j.ygyno.2019.11.0130090-8258/© 2019 Elsevier Inc. All rights reserved.

a b s t r a c t

Objective: High-grade serous ovarian cancers (HGSOC) are genomically characterized by homologousrecombination deficiency (HRD) and TP53 mutations, which lead to intratumor heterogeneity (ITH). Thisstudy aimed to reveal the relationship between HRD, ITH and prognosis and analyze their changes duringtreatment.Methods: We obtained 573 SNP array and gene expression array data from The Cancer Genome Atlas.SNP array data were processed to calculate the Clonality Index (CI) and loss of heterozygosity (LOH)scores. Gene expression array data were used for classifying molecular subtypes. Additionally, we ob-tained 33 samples from 20 HGSOC patients, including 4 samples from interval debulking surgery (IDS)and 9 samples from recurrent surgery.Results: We divided HGSOC samples into 2 groups. The high CI group showed a high recurrent risk, andthe high LOH group showed a statistically good prognosis. Combining the two factors, the high LOH/lowCI group showed a statistically good prognosis. In terms of molecular subtypes, the mesenchymal sub-type, which had a poor prognosis, showed a high CI with statisitically significant difference and theimmunoreactive subtype, which had a good prognosis, showed a tendency to have a high LOH score.Throughout treatment, the CI decreased to one at the IDS (n ¼ 4) and then increased at recurrence(n ¼ 3). LOH scores greatly decreased in two cases at the IDS.Conclusions: ITH and HRD were associated with prognosis in HGSOC. ITH decreased after neoadjuvantchemotherapy, suggesting that the chemo-resistant cancer clone remains after chemotherapy.

© 2019 Elsevier Inc. All rights reserved.

n cancer; HRD, homologouse Atlas; LOH, loss of hetero-therapy.

aya), [email protected]), [email protected]), [email protected]).

1. Introduction

Ovarian cancer has a poor prognosis among gynecologic ma-lignancies [1]. High-grade serous ovarian cancer (HGSOC), which isthemost frequent epithelial ovarian cancer, is commonly diagnosedas an advanced cancer [2]. HGSOC is mainly treated by chemo-therapy; however, most HGSOCs recur as a chemo-resistant tumor

Page 2: Intratumor heterogeneity and homologous recombination ...

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422416

even though the first chemotherapy is usually effective and resultsin lethal treatment.

The Cancer Genome Atlas (TCGA) has revealed some charac-teristics of the cancer genome of HGSOC. One of the characteristicsis homologous recombination repair deficiency (HRD) [3]. Almosthalf of HGSOCs have HRD due to BRCA1/2 mutation or other ho-mologous recombination repair pathway-related gene alterations[4]. HRD is associated with sensitivity to platinum agents [5] andpoly- (ADP ribose) polymerase (PARP) inhibitors [6e8], and quan-tifying the genomic loss of heterozygosity (LOH) provides thepossibility of measuring HRD as a biomarker [9]. In the ARIEL2study, the LOH score was used as a treatment biomarker for thePARP inhibitor, and it was shown that the PARP inhibitor waseffective in cases with a high LOH score [10].

The TP53 mutation can be detected at a high frequency inHGSOC [3]. TP53mutations give rise to chromosomal instability dueto disruption of the control of the cell cycle and apoptosis [11].Chromosomal instability causes subclonal evolution, which showsdifferent genomic characteristics in tumorigenesis and tumorprogression and results in high intratumor heterogeneity [12,13],which is associated with resistance to treatment and poor prog-nosis [14e16]. We have already reported that a single nucleotidepolymorphism (SNP) array with formalin-fixed paraffin-embeddedspecimens can be used to analyze intratumor heterogeneity [17].With this method, we can evaluate intratumor heterogeneity fromvarious clinical specimens and apply the findings to clinical fea-tures, such as prediction of prognosis.

Although each of these main characteristics of HGSOC wasinvestigated in several studies, no reports have examined the as-sociation between HRD and intratumor heterogeneity and howthese factors changed before and after chemotherapy. In the cur-rent study, we first investigated the association between prognosisand HRD or intratumor heterogeneity with the use of TCGA data,and second, we analyzed the changes in these factors throughouttreatment of HGSOC with samples from our institution.

2. Materials and methods

2.1. Data sources

We obtained TCGA CEL-formatted SNP array data from Affy-metrix Genome-Wide Human SNP Array 6.0 via the Genomic DataCommons (GDC) Data Portal (https://portal.gdc.cancer.gov) andextracted 573 HGSOC cases with matched tumor-normal data. Weobtained TCGA CEL-formatted gene expression array data from theGeneChip HT Human Genome U133A data via the GDC Data Portaland extracted cases having the same samples as the SNP array data.Additionally, we obtained TCGA XML-formatted clinical data foreach case via the GDC Data Portal.

2.2. Clonality Index (CI) estimation

We previously reported a method to estimate the clonalcomposition using the OncoScan FFPE Assay Kit [17]. The logR ratio(LRR) and B-allele frequency (BAF) can be calculated with the copynumber of tumor cells, proportion of aberrant cells (%AC), and copynumber of minor alleles (NOMA), theoretically. When we plot theLRR and logarithm of BAF in 2-dimensional space, if the %AC is thesame and NOMA equals zero, the plot makes a straight line.Therefore, we can calculate the %AC from the observed LRR and BAFif the NOMA equals zero in a segment, and we can estimate thenumber of clones of the tumor as the CI by analyzing the distri-bution of a set of the %AC.

We created an algorithm for calculating the CI under the envi-ronment of Python 3.6 and R version 3.5.0 as described above. Thesegmentation data of the LRR and BAF were obtained from CEL-

formatted SNP array data using the R package “rawcopy” [18]. Weexcluded segmentation datawhose base pair length was equal to orless than the threshold (default settings ¼ 1000 base pairs). Wecalculated the weighted Euclidean distance between the co-ordinates of the observed LRR/observed BAF and the theoreticalLRR/theoretical BAF in each %AC, and then the %AC at which thedistance was minimized and less than 0.1 was determined as the %AC for that segment. If the number of output %AC was one or less,the CI was not unable to be estimated. Because the distribution of aset of %AC in the samples was unimodal or multimodal, we couldestimate the most appropriate shape of the distribution thatminimized the Bayesian information criterion using the R package“Mclust”, and the number of clusters of the distribution was takenas the CI. If a cluster included less than 1% of the total %AC, thatcluster was excluded.

2.3. LOH score calculation

CEL-formatted SNP array files were processed by AffymetrixPower Tools and PennCNV software [19] to calculate the LRR andBAF data in each probe, and then segmented copy number datawere analyzed using the R package “ASCAT” [20]. The LOH scorewas calculated as the proportion of the sum of the LOH regionwhose length was equal to or greater than 15 M base pairs to thelength of all chromosomes, as previously reported [10,21]. Weexcluded the LOH region, which was more than 90% of the shortarm, long arm, or total chromosome, in each chromosome.

2.4. Analysis of gene expression array data

CEL-formatted gene expression array data were normalized bythe Robust Multiarray Average (RMA) method using the R package“aroma.light”. Single sample gene set enrichment analysis (ssGSEA)was performed to classify HGSOC samples into four gene expres-sion subtypes as previously defined [22]. Differential gene expres-sion analysis was performed by Welch’s t-test, and p-values werecorrected by the Benjamini-Hochberg method. Pathway analysiswas performed by Ingenuity Pathway Analysis (IPA; QIAGEN).

2.5. Clinical data extraction

XML-formatted clinical data files were read using Python, andthe clinical information was extracted, including age at diagnosis,the International Federation of Gynecology and Obstetrics (FIGO)stage, treatment of neoadjuvant chemotherapy (NAC), site ofanalyzed tumor, residual tumor size, primary outcome, survivaldata, and vital status.

2.6. Analysis of our HGSOC samples

Thirty-three tumor samples from 20 HGSOC cases who under-went primary surgery between 1994 and 2012 in Kindai UniversityHospital, including 20 samples from primary surgery (14 samplesfrom ovary, 4 samples from peritoneal metastasis, and 2 samplesfrom omentum), 4 samples from interval debulking surgery (IDS)after NAC (3 samples from ovary and 1 sample from omentum), and9 samples from recurrent surgery (7 samples from peritonealmetastasis and 1 sample from lymph node and brain metastasis),were retrospectively analyzed. The pathological diagnosis ofHGSOC was diagnosed at the Central Pathological Department inKindai University Hospital using either hematoxylin and eosin(HE)-stained slides or additional immunohistochemistry analysis,the results of which were reviewed by at least one gynecologiconcologist. The formalin-fixed, paraffin-embedded (FFPE) tumorspecimens stained with HE were reviewed to confirm the presenceof more than 50% of viable tumor cells by at least one author

Page 3: Intratumor heterogeneity and homologous recombination ...

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422 417

specializing in gynecologic pathology and oncology after theconfirmation of the Central Pathological Department, and the tu-mor regions were removed manually. Genomic DNA was extractedfrom FFPE specimens using an AllPrep DNA/RNA FFPE Kit (QIAGEN).The quality of DNA was analyzed using a NanoDrop 2000/2000cSpectrophotometer (ThermoFisher Scientific), and the quantity ofDNA was measured using a Quant-iT PicoGreen dsDNA Assay Kit(ThermoFisher Scientific).

DNA extracted from FFPE specimens was analyzed with anOncoScan FFPE Assay Kit (ThermoFisher Scientific). CEL-formattedfiles were output with Affymetrix GeneChip Command Consolesoftware version 4.0 and converted to OSCHP files with OncoScanConsole software 1.3. The number of clones was estimated with theOncoClone Composition program [17] using OSCHP files. OSCHPfiles including the LRR and BAF data in each probewere analyzed byASCAT to generate copy number segmented data, and the LOH scorewas calculated. Samples with aberrant cell fractions less than 30%were excluded as previously reported [23]. GISTIC 2.0 [24] was usedto identify copy number aberrations in tumors.

2.7. Statistical analysis

The correlation analysis between the CI and the LOH score wasperformed with Spearman correlation coefficient. The comparisonbetween molecular subtypes was performed with the Kruskal-Wallis test. The CI was dichotomized based on a median split asperformed in previous studies, where authors performed a mediansplit to dichotomize samples into two subgroups [25e27] to illus-trate the association between the CI and survival time. The LOHscore was also dichotomized based on the Gaussian mixture model.The survival curves were described with Kaplan-Meier estimation,and survival analysis was performed with the log-rank test. Toconsider confounding factors with respect to the survival analysis,covariates such as age at diagnosis (as a continuous variable), FIGOstage (I/II vs III/IV), and residual tumor size (nomacroscopic diseasevs 1e10 mm vs > 10 mm) were applied to the Cox proportional

Fig. 1. Histogram and boxplot of Clonality Index (CI) (A) and LOH scores (B) and pair plot of Cwas 17.9, and the median was 17.26. Correlation between CI and LOH score. The Spearman

hazards model along with the CI and the LOH score to examine theassociation with the CI or the LOH score on survival time afteradjusting these covariates. All statistical tests were performed withR, and a p-value of <0.05 was considered statistically significant.

3. Results

CI and LOH scores could be estimated in 536 and 542 samplesout of 573 samples, respectively. Both CI and LOH scores could becalculated in 502 samples, so 502 samples were subjected toanalysis. The background data for patients in each sample set areshown in Supplementary Table 1. The averages and standard de-viations of the CI and LOH scores were 3.53 ± 1.95 and 17.90 ± 9.41,respectively (Fig. 1A and B). The correlation coefficient between theCI and LOH scores was �0.176, which indicated a slight negativecorrelation (Fig. 1C).

We divided samples into 2 groups based on a median split: ahigh CI group, which had scores of three or higher, and a low CIgroup, which had scores lower than 3. In the survival analysis, themedian progression-free survival (PFS) of the high and low CIgroups was 16.6 months vs 20.6 months (p-value ¼ 0.0097),respectively, with a significant difference observed. However, themedian overall survival (OS) in the high and low CI groups was 44.5months and 48.2 months (p-value ¼ 0.419), respectively (Fig. 2Aand B).

We set a cut-off value of 16 for the LOH score based on theGaussianmixturemodel and divided the samples into 2 groups: thehigh LOH group, which had scores of 16 or more, and the low LOHgroup, which had scores lower than 16. In the survival analysis, themedian PFS and OS in the high LOH and low LOH groups was 19.3months vs 16.3 months (p-value¼ 0.0056) and 50.0 months vs 38.6months (p-value < 0.001), respectively, with significant differencesobserved (Fig. 2C and D).

In the Cox proportional hazards model adjusting for age, FIGOstage, and residual tumor size, high CI was independently associ-ated with a shorter PFS (adjusting hazard ratio (HR) ¼ 1.473, 95%

I and LOH scores (C). The mean CI was 3.53, and the median was 3. The mean LOH scorecorrelation coefficient for the CI and LOH scores was �0.176.

Page 4: Intratumor heterogeneity and homologous recombination ...

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422418

CI: ¼ 1.140e1.904, p-value ¼ 0.003, Supplementary Table 2) andhigh LOH scorewas independently associatedwith a longer PFS andOS (adjusting HR ¼ 0.698 and 0.644, 95% CI ¼ 0.538e0.905 and0.501e0.828, p-value ¼ 0.007 and <0.001, respectively,Supplementary Table 3). When both the CI and the LOH score weresimultaneously included in the Cox model with the covariates, theCI remained significantly associated with a shorter PFS (adjustingHR ¼ 1.392, 95% CI ¼ 1.072e1.808, p-value ¼ 0.0132), and the LOHscore remained indicative of a better prognosis (OS: adjustingHR ¼ 0.648, 95% CI ¼ 0.502e0.834, p-value < 0.001, PFS: adjustingHR ¼ 0.747, 95% CI ¼ 0.574e0.973, p-value ¼ 0.031).

We stratified samples into 4 groups based on the combination ofthe CI and LOH scores: a high LOH/high CI group, a high LOH/low CIgroup, a low LOH/high CI group, and a low LOH/group LOH group.

Fig. 2. Kaplan-Meier curve of TCGA data stratified with the Clonality Index (CI) value (A, B),progression-free survival (PFS) (B, D, F). The low CI group showed significantly prolongeddifferences. The combined high LOH and low CI group showed good prognosis compared w

We subsequently performed a survival analysis. Comparing thehigh LOH/low CI group with the high LOH/high CI, low LOH/high CI,and low LOH/low CI groups, the median PFS was 22.6 months vs16.8, 16.6, and 15.0 months (p-value < 0.001), and the median OSwas 55.1 months vs 49.5, 38.6, and 38.6 months (p-value < 0.001),respectively. There were statistically significant differences (Fig. 2Eand F).

Five hundred and twenty-two gene expression array data fromthe TCGA database were subject to ssGSEA with the CLOVAR geneset to classify 4 molecular subtypes: differentiated, immunoreac-tive, mesenchymal, and proliferative. Subsequently, 454 sampleswere analyzed based on the CI, LOH score, and molecular subtype.The average CI values in differentiated, immunoreactive, mesen-chymal, and proliferative subtypes were 3.26, 3.69, 3.85, and 3.52,

LOH score (C, D), and a combination of both (E, F) for overall survival (OS) (A, C, E) andPFS but not OS. The high LOH group showed prolonged OS and PFS with significantith the other groups.

Page 5: Intratumor heterogeneity and homologous recombination ...

Fig. 3. Pair plot and boxplot between each gene expression subtype and CI (A) and LOH scores (B). The mesenchymal subtype had a higher CI than the other subtypes, and theimmunoreactive subtype had the highest LOH score.

Table 1Patient characteristics.

(N ¼ 20)

Age, median(range) 56(42e82)FIGO stageI/II 2III/IV 18

Primary surgeryMaximum cytoreductive 14Probe laparotomy/laparoscopy 6

Size of residual tumorsNo macroscopic disease 71e10 mm 8>10 mm 5

Sites of recurrencePeritoneal dissemination 7Lymph node 1Organ metastasis 2

FIGO; International Federation of Gynecology and Obstetrics.

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422 419

respectively. The mesenchymal subtype, which had a poor prog-nosis, showed a high CI (Fig. 3A, p-value ¼ 0.0403). The averageLOH scores in each subtype were 17.5, 19.2, 17.3, and 16.2, respec-tively. There was no significant difference in the LOH score betweenmolecular subtypes, but the immunoreactive subtype, which had agood prognosis, showed a tendency to have a high LOH score(Fig. 3B, p-value ¼ 0.0762).

We performed downstream effect analysis with IPA comparinghigh CI and low CI, high LOH and low LOH, and high LOH and low CIand other groups (Supplementary Tables 5e7). In the high LOHgroup and high LOH/low CI group, it was predicted that the acti-vation, migration, and adhesion of immune cells were activated.

Next, we analyzed 33 HGSOC samples from 20 cases from KindaiUniversity Hospital. Patient characteristics are shown in Table 1.Wedivided 20 samples before chemotherapy into a high CI group (CIwas 3 or more) and a low CI group (CI less than 3) and performedsurvival analysis. The low CI group showed a significantly longerPFS and OS (Supplementary Fig. 1, p-value ¼ 0.0351, 0.0385,respectively). In the Cox proportional hazardsmodel with adjustingfor clinical information, a high CI value was associated with ashorter PFS (adjusting HR ¼ 8.078, 95% CI: ¼ 1.117e58.413, p-value¼ 0.0385, Supplementary Table 4). We analyzed the change inthe CI throughout the time of treatment in 4 cases that could beanalyzed for tumor samples at IDS after NAC, including one casethat could not be analyzed at recurrence. The CI at primary surgerywas 3 in 3 cases and 2 in 1 case, and at IDS, the CI decreased to 1 inall 4 cases, and then the CI increased at the recurrent surgery in all 3cases (Fig. 4A). It was found that the tumor clone consisted of

multiple clones before chemotherapy. However, after chemo-therapy, the chemo-sensitive clone decreased and appeared todecrease the CI, and at the time of recurrence, the tumor cloneincreased again. We analyzed changes in the LOH score in the sameway, and the LOH score did not change in 2 cases (30.2 / 29.2,27.3/ 25.4) and greatly decreased in 2 cases from primary surgeryto IDS and then increased at recurrent surgery (15.7 / 5.7 / 16.5,28.6 / 12.0 / 30.0) (Fig. 4B). In 6 cases whose samples wereanalyzed at primary and recurrent surgery, CI did not change in 4cases and increased and decreased in one case each. The LOH scoredid not change in 5 cases and decreased in one case (Fig. 4C and D).With GISTIC analysis, a pattern of the copy number variantcompared between primary and recurrent surgery had few differ-ences, but amplification of 8q24 was found at IDS with statisticalsignificance (Fig. 5AeC).

4. Discussion

We analyzed intratumor heterogeneity and HRD in HGSOC withSNP array data in the current study. Some methods to analyze theintratumor heterogeneity of malignant tumors, including ovariancancer, have previously been reported, such as multi-regionsequencing [28], deep sequencing [29], and single-cell sequencing[30]. We could analyze intratumor heterogeneity from a singlesampling of a tumor by applying a previous method [17], which wereported in the current study. The merit of this method is as fol-lows: 1) public data such as TCGA data can be applied, 2) residualtumors after chemotherapy can be analyzed as FFPE tumor speci-mens, and 3) easy application in clinical settings is possible becausethe cost of analysis is relatively low compared with that ofsequencing methods.

In this study, we report two findings regarding the associationbetween intratumor heterogeneity and the prognosis of HGSOC:PFS in the high intratumor heterogeneity group was shorter thanthat observed in the low intratumor heterogeneity group as anindependent factor, and there was no difference in OS with respectto intratumor heterogeneity. Several studies have reported that ahigh degree of intratumor heterogeneity-induced resistance totreatment leads to poor prognosis in various types of cancer. Ohet al. reported that tumors with high intratumor heterogeneity ofcolorectal cancer had a shorter PFS [16]. Andor et al. analyzed therelationship between the number of clones and prognosis using 12types of cancer with TCGA data and reported that a high clonenumber was related to poor prognosis across cancer types [31].Morris et al. analyzed the relationship between intratumor het-erogeneity and prognosis using 9 types of cancer with TCGA data

Page 6: Intratumor heterogeneity and homologous recombination ...

Fig. 4. The transition of CI and LOH scores throughout the treatment. The CI was reduced to 1 at interval debulking surgery (IDS) and increased at recurrence in all 4 cases (A). TheLOH score was prominently reduced at IDS and increased at recurrence in 2 cases, but in the other 2 cases, the LOH score was not changed (B). In 6 cases without IDS data, the CI didnot change in 4 cases (C), and LOH scores were almost the same (D).

Fig. 5. Amplification of HGSOC samples at primary surgery (A), interval debulking surgery (B), and recurrent surgery (C) from GISTIC analysis. Chromosome 8q24 was amplified atall three surgical times.

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422420

and reported high intratumor heterogeneity related to shorter OSin several types of cancer [14]. In the current study, our results onPFS were similar with those of previous reports, although thefinding regarding the OS was different from that observed in pre-vious studies. However, pan-cancer studies reported by Andor et al.

and Morris et al. did not include ovarian cancer, so it is difficult todirectly compare their reports with our study. Moreover, Andoret al. revealed the association between OS and the number of clonesacross all cancer types but not individual cancer type. Additionally,the association between intratumor heterogeneity and OS was not

Page 7: Intratumor heterogeneity and homologous recombination ...

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422 421

shown in bladder cancer, lung adenocarcinoma, and lung squamouscell carcinoma in the study from Morris et al.. Thus, from theseprevious studies, the association between intratumor heterogene-ity and OS might be different by cancer type.

HRD related to platinum sensitivity and prognosis, and theproportion of the LOH could be a biomarker of HRD [5,32,33]. In thecurrent study, HGSOC cases with high LOH scores were associatedwith a good prognosis (Fig. 2C and D) as an independent factor.Furthermore, molecular subtypes of HGSOC had different progno-ses [22]; the CI was high in the mesenchymal subtype, which had apoor prognosis, and LOH tended to be high in the immunoreactivesubtype, which had a good prognosis (Fig. 3A and B). Additionally,the pathways related to immune function were upregulated incases with high LOH scores (Supplementary Tables 6 and 7).Consequently, intratumor heterogeneity and HRD status arepartially related to differences in prognosis between molecularsubtypes. In a previous study, cases in the immunoreactive subtypehad a high frequency of BRCA1 mutations [34].

This study is the first to analyze the changes in intratumorheterogeneity before and after NAC in HGSOC.We revealed that theCI of the tumor that remained at the IDS decreased; hence, chemo-sensitive clones are thought to disappear and chemo-resistantclones are thought to remain after chemotherapy. Additionally,chromosome 8q24 amplification was detected in the tumorsremaining after chemotherapy (Fig. 5B). Several tumor-relatedgenes, such as MYC, PVT-1, PSCA, and TNFRSF11B, were located onchromosome 8q24, and amplification of this region was related topoor prognosis in gastric cancer, breast cancer, colorectal cancer,and ovarian cancer in a previous study [35e38]. The LOH scoreprominently decreased after chemotherapy in two cases (Fig. 4B).This result indicated that intratumor heterogeneity of HRD statusdecreased after chemotherapy, that is, non-HRD clones wereselected. On the other hand, in another two cases, the LOH score didnot decrease after chemotherapy. Although these cases had highLOH scores, which represent HRD, the LOH score did not change;hence, this result showed that the chemosensitivity was not alwaysconsistent with HRD status.We analyzed only 4 cases in the currentstudy, so we need to increase the number of cases in futureanalyses.

Comparing the tumor at primary and recurrent surgery, therewere few changes in the CI, LOH score, and patterns of copy numbervariation. In a previous study, mutation status or copy numbervariation detected by target sequencing [29] or whole exomesequencing [39] drastically changed between primary and recur-rent tumors; however, one study reported that HRD status did notchange [39]. The current study indicated that the tumor thatremained after chemotherapywas very different from the recurrenttumor. Therefore, we should analyze the tumor that remained afterchemotherapy precisely to investigate the mechanism of chemo-resistance.

There are some limitations of this study. First, the methoddeveloped to calculate the CI in this study cannot be applied to anycancer, since its use depends on amount of LOH region of the cancergenome. When a cancer has few copy number variations, the CI ofthe cancer can be underestimated. Second, the number of sampleswas limited in our own data. Both the total number of cases andNAC samples was inadequate to draw any statistically meaningfulconclusions. Thus, further studies with additional samples areneeded to confirm the current conclusion. Third, when wecompared the CI values and the LOH scores before and afterchemotherapy, the analyzed tumor tissue was obtained fromdifferent site of lesions in the same case. Because genetic orphenotypic variations between tumors would occur at differentsites in the same patients, which is known to be intertumoralheterogeneity [40], comparing the CI values of different tumorsbefore and after chemotherapy in the same patients many not

adequately allow the effect of chemotherapy on the tumors to beevaluated.

5. Conclusion

We have shown that both the CI and LOH in HGSOC is related toprognosis using SNP array data from a single sample. Furthermore,we have shown that the intratumor heterogeneity of chemo-resistant tumors that remained at IDS after NAC decreasedcompared with that of primary tumors. This study indicated thatfor analysis of tumors that remain after chemotherapy, investiga-tion of the mechanism underlying the development of chemo-resistance is important; thus, further studies are needed.

Declaration of competing interest

All authors declare no conflicts of interest.

Acknowledgements

This work was supported by JSPS Grant-in-Aid for Young Sci-entists (B) Grant Number JP17K16873.

Appendix A. Supplementary data

Supplementary data to this article can be found online athttps://doi.org/10.1016/j.ygyno.2019.11.013.

References

[1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2019, CA A Cancer J. Clin. 69(2019) 7e34, https://doi.org/10.3322/caac.21551.

[2] J.D. Seidman, I. Horkayne-Szakaly, M. Haiba, et al., The histologic type andstage distribution of ovarian carcinomas of surface epithelial origin, Int. J.Gynecol. Pathol. 23 (2004) 41e44, https://doi.org/10.1097/01.pgp.0000101080.35393.16.

[3] Cancer GenomeAtlas ResearchNetwork, Integrated genomic analyses of ovariancarcinoma, Nature 474 (2011) 609e615, https://doi.org/10.1038/nature10166.

[4] P.A. Konstantinopoulos, R. Ceccaldi, G.I. Shapiro, A.D. D???Andrea, Homolo-gous recombination deficiency: exploiting the fundamental vulnerability ofovarian cancer, Cancer Discov. 5 (2015) 1137e1154, https://doi.org/10.1158/2159-8290.CD-15-0714.

[5] E.Y. Zhao, Y. Shen, E. Pleasance, et al., Homologous recombination deficiencyand platinum-based therapy outcomes in advanced breast cancer, Clin. CancerRes. 23 (2017) 7521e7530, https://doi.org/10.1158/1078-0432.CCR-17-1941.

[6] J. Ledermann, P. Harter, C. Gourley, et al., Olaparib maintenance therapy inpatients with platinum-sensitive relapsed serous ovarian cancer: a pre-planned retrospective analysis of outcomes by BRCA status in a randomisedphase 2 trial, Lancet Oncol. 15 (2014) 852e861, https://doi.org/10.1016/S1470-2045(14)70228-1.

[7] K.A. Gelmon, M. Tischkowitz, H. Mackay, et al., Olaparib in patients withrecurrent high-grade serous or poorly differentiated ovarian carcinoma ortriple-negative breast cancer: a phase 2, multicentre, open-label, non-rand-omised study, Lancet Oncol. 12 (2011) 852e861, https://doi.org/10.1016/S1470-2045(11)70214-5.

[8] J. Mateo, S. Carreira, S. Sandhu, et al., DNA-repair defects and olaparib inmetastatic prostate cancer, N. Engl. J. Med. 373 (2015) 1697e1708, https://doi.org/10.1056/NEJMoa1506859.

[9] V. Abkevich, K.M. Timms, B.T. Hennessy, et al., Patterns of genomic loss ofheterozygosity predict homologous recombination repair defects in epithelialovarian cancer, Br. J. Canc. 107 (2012) 1776e1782, https://doi.org/10.1038/bjc.2012.451.

[10] E.M. Swisher, K.K. Lin, A.M. Oza, et al., Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international,multicentre, open-label, phase 2 trial, Lancet Oncol. 18 (2017) 75e87, https://doi.org/10.1016/S1470-2045(16)30559-9.

[11] W. Hanel, U.M. Moll, Links between mutant p53 and genomic instability,J. Cell. Biochem. 113 (2012) 433e439, https://doi.org/10.1002/jcb.23400.

[12] S.F. Bakhoum, L.C. Cantley, The multifaceted role of chromosomal instability incancer and its microenvironment, Cell 174 (2018) 1347e1360, https://doi.org/10.1016/j.cell.2018.08.027.

[13] L. Sansregret, B. Vanhaesebroeck, C. Swanton, Determinants and clinical im-plications of chromosomal instability in cancer, Nat. Rev. Clin. Oncol. 15(2018) 139e150, https://doi.org/10.1038/nrclinonc.2017.198.

[14] L.G.T. Morris, N. Riaz, A. Desrichard, et al., Pan-cancer analysis of intratumorheterogeneity as a prognostic determinant of survival, Oncotarget 7 (2016)10051e10063, https://doi.org/10.18632/oncotarget.7067.

Page 8: Intratumor heterogeneity and homologous recombination ...

H. Takaya et al. / Gynecologic Oncology 156 (2020) 415e422422

[15] J. Oltmann, K. Heselmeyer-Haddad, L.S. Hernandez, et al., Aneuploidy, TP53mutation, and amplification of MYC correlate with increased intratumorheterogeneity and poor prognosis of breast cancer patients, Genes Chromo-somes Cancer 57 (2018) 165e175, https://doi.org/10.1002/gcc.22515.

[16] B.Y. Oh, H.-T. Shin, J.W. Yun, et al., Intratumor heterogeneity inferred fromtargeted deep sequencing as a prognostic indicator, Sci. Rep. 9 (2019) 4542,https://doi.org/10.1038/s41598-019-41098-0.

[17] K. Sakai, M. Ukita, J. Schmidt, et al., Clonal composition of human ovariancancer based on copy number analysis reveals a reciprocal relation withoncogenic mutation status, Cancer Lett. 405 (2017) 22e28, https://doi.org/10.1016/j.canlet.2017.07.013.

[18] M. Mayrhofer, B. Viklund, A. Isaksson, Rawcopy: improved copy numberanalysis with Affymetrix arrays, Sci. Rep. 6 (2016) 36158, https://doi.org/10.1038/srep36158.

[19] K. Wang, M. Li, D. Hadley, et al., PennCNV: an integrated hidden Markovmodel designed for high-resolution copy number variation detection inwhole-genome SNP genotyping data, Genome Res. 17 (2007) 1665e1674,https://doi.org/10.1101/gr.6861907.

[20] P. Van Loo, S.H. Nordgard, O.C. Lingjaerde, et al., Allele-specific copy numberanalysis of tumors, Proc. Natl. Acad. Sci. 107 (2010) 16910e16915, https://doi.org/10.1073/pnas.1009843107.

[21] R.L. Coleman, A.M. Oza, D. Lorusso, et al., Rucaparib maintenance treatmentfor recurrent ovarian carcinoma after response to platinum therapy (ARIEL3):a randomised, double-blind, placebo-controlled, phase 3 trial, Lancet 390(2017) 1949e1961, https://doi.org/10.1016/S0140-6736(17)32440-6.

[22] R.G.W. Verhaak, P. Tamayo, J.-Y. Yang, et al., Prognostically relevant genesignatures of high-grade serous ovarian carcinoma, J. Clin. Investig. 123 (2012)517e525, https://doi.org/10.1172/JCI65833.

[23] A.M. Marquard, A.C. Eklund, T. Joshi, et al., Pan-cancer analysis of genomic scarsignatures associated with homologous recombination deficiency suggestsnovel indications for existing cancer drugs, Biomark. Res. 3 (2015) 9, https://doi.org/10.1186/s40364-015-0033-4.

[24] C.H. Mermel, S.E. Schumacher, B. Hill, et al., GISTIC2.0 facilitates sensitive andconfident localization of the targets of focal somatic copy-number alterationin human cancers, Genome Biol. 12 (2011) R41, https://doi.org/10.1186/gb-2011-12-4-r41.

[25] R. Stickgold, J.A. Hobson, R. Fosse, M. Fosse, Sleep, learning, and dreams: off-line memory reprocessing, Science 294 (2001) 1052e1057, https://doi.org/10.1126/science.1063530.

[26] D.G. Rand, J.D. Greene, M.A. Nowak, Spontaneous giving and calculated greed,Nature 489 (2012) 427e430, https://doi.org/10.1038/nature11467.

[27] M. Jamal-Hanjani, G.A. Wilson, N. McGranahan, et al., Tracking the evolutionof nonesmall-cell lung cancer, N. Engl. J. Med. NEJMoa1616288 (2017),https://doi.org/10.1056/NEJMoa1616288.

[28] A. Bashashati, G. Ha, A. Tone, et al., Distinct evolutionary trajectories of pri-mary high-grade serous ovarian cancers revealed through spatial mutationalprofiling, J. Pathol. 231 (2013) 21e34, https://doi.org/10.1002/path.4230.

[29] L. Beltrame, M. Di Marino, R. Fruscio, et al., Profiling cancer gene mutations inlongitudinal epithelial ovarian cancer biopsies by targeted next-generationsequencing: a retrospective study, Ann. Oncol. 26 (2015) 1363e1371,https://doi.org/10.1093/annonc/mdv164.

[30] A. McPherson, A. Roth, E. Laks, et al., Divergent modes of clonal spread andintraperitoneal mixing in high-grade serous ovarian cancer, Nat. Genet. 48(2016) 758e767, https://doi.org/10.1038/ng.3573.

[31] N. Andor, T.A. Graham, M. Jansen, et al., Pan-cancer analysis of the extent andconsequences of intratumor heterogeneity, Nat. Med. 22 (2016) 105e113,https://doi.org/10.1038/nm.3984.

[32] M. Tumiati, S. Hietanen, J. Hynninen, et al., A functional homologous recom-bination Assay predicts primary chemotherapy response and long-term sur-vival in ovarian cancer patients, Clin. Cancer Res. 24 (2018) 4482e4493,https://doi.org/10.1158/1078-0432.CCR-17-3770.

[33] B.M. Norquist, M.F. Brady, M.I. Harrell, et al., Mutations in homologousrecombination genes and outcomes in ovarian carcinoma patients in GOG218: an NRG oncology/Gynecologic oncology group study, Clin. Cancer Res. 24(2018) 777e783, https://doi.org/10.1158/1078-0432.CCR-17-1327.

[34] J. George, K. Alsop, D. Etemadmoghadam, et al., Nonequivalent gene expres-sion and copy number alterations in high-grade serous ovarian cancers withBRCA1 and BRCA2 mutations, Clin. Cancer Res. 19 (2013) 3474e3484, https://doi.org/10.1158/1078-0432.CCR-13-0066.

[35] F. Meng, B. Liu, G. Xie, et al., Amplification and overexpression of PSCA at 8q24in invasive micropapillary carcinoma of breast, Breast Canc. Res. Treat. 166(2017) 383e392, https://doi.org/10.1007/s10549-017-4407-1.

[36] X. Wang, Y. Liu, D. Shao, et al., Recurrent amplification of MYC and TNFRSF11Bin 8q24 is associated with poor survival in patients with gastric cancer, GastricCancer 19 (2016) 116e127, https://doi.org/10.1007/s10120-015-0467-2.

[37] Y. Takahashi, G. Sawada, J. Kurashige, et al., Amplification of PVT-1 is involvedin poor prognosis via apoptosis inhibition in colorectal cancers, Br. J. Canc. 110(2014) 164e171, https://doi.org/10.1038/bjc.2013.698.

[38] Y. Guan, W.-L. Kuo, J.L. Stilwell, et al., Amplification of PVT1 contributes to thepathophysiology of ovarian and breast cancer, Clin. Cancer Res. 13 (2007)5745e5755, https://doi.org/10.1158/1078-0432.CCR-06-2882.

[39] S. Lambrechts, D. Smeets, M. Moisse, et al., Genetic heterogeneity after first-line chemotherapy in high-grade serous ovarian cancer, Eur. J. Cancer 53(2016) 51e64, https://doi.org/10.1016/j.ejca.2015.11.001.

[40] R.A. Burrell, N. McGranahan, J. Bartek, C. Swanton, The causes and conse-quences of genetic heterogeneity in cancer evolution, Nature 501 (2013)338e345, https://doi.org/10.1038/nature12625.