Open Access

Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer

  • Authors:
    • Xiaoxiang Jie
    • Ming Du
    • Meng Zhang
    • Xiayu Jin
    • Qingqing Cai
    • Congjian Xu
    • Xiaoyan Zhang
  • View Affiliations

  • Published online on: June 30, 2022     https://doi.org/10.3892/etm.2022.11479
  • Article Number: 542
  • Copyright: © Jie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Circulating tumor DNA (ctDNA) is one conventional type of liquid biopsy that can be collected to dynamically monitor disease status. However, its potential clinical value and concordance with ascites samples or tumor biopsy needs to be evaluated further for patients with ovarian cancer. Therefore, the present study compared the mutation profiles among ctDNA, paired tumor tissue and ascites samples to explore their possible clinical value in ovarian cancer. Targeted next‑generation sequencing was used to screen for mutations in 18 peripheral blood samples, six paired ascites samples and eight paired tumor tissues collected from patients with ovarian cancer. Functional analyses were performed using public databases. WebGestalt was used to perform Gene Ontology and pathway enrichment analyses. The cBioPortal for Cancer Genomics was used to assess therapeutic targets. Chilibot and Search Tool for the Retrieval of Interacting Genes/Proteins were used to obtain key genes and their functional interactions. Comparative analysis was performed among the three types of samples using Venn diagram. A total of 104 cancer‑associated mutant genes in ctDNA samples, 95 genes in tumor tissues and 44 genes in ascites samples were found. A cluster covering 10 genes, namely NOTCH2, NOTCH3, lysine methyltransferase 2A, PTEN, androgen receptor, DNA‑activated protein kinase catalytic subunit, hepatocyte nuclear factor 1 homeobox A, SRC, insulin receptor substrate 2 and SRY‑box transcription factor 10, was obtained by Chilibot analysis. This gene panel may have the potential to monitor metastasis and identify therapeutic targets in ovarian cancer. Taken together, the present study focused on the mutant genes in ctDNA, ascites and tumor tissues, and suggested that the integrated information of different samples could be examined to comprehensively reflect the mutational landscape in ovarian cancer. However, procedures and protocols to interpret and utilize the integrated information obtained from various forms of liquid biopsies will require optimization prior to their use for future clinical applications.

Introduction

According to cancer statistics of 2022, ovarian cancer ranks as the fifth major cause of cancer-associated mortality for females in the United States, with a 5-year survival rate <50% for all stages (1). In addition, ~60% of newly diagnosed patients typically present with distant metastasis, resulting in a 30% 5-year survival rate (2). Due to the anatomical location of the ovaries in the deep pelvic cavity, symptoms of this cancer include abdominal or pelvic pain or distension, bloating, urinary frequency and urgency (3,4). However, they are only perceptible after metastatic malignancy has been developed or after the volume of the primary tumor has reached a size that can obstruct the gastrointestinal or pelvic organs (3,4). Genetic heterogeneity is widespread in ovarian cancer (5-7). In ovarian cancer, genomic variation, invasive ability and sensitivity to platinum profiles are all heterogeneous among intratumor subclones, subtypes or between primary and recurrent malignancy (8-10). This poses a challenge to the development of applicable genetic biomarkers for early diagnosis, dynamic disease monitoring and therapeutic target identification in the clinic. Identification of novel biomarkers and deeper understanding of the genomic profile of ovarian cancer will likely increase the survival rate from this disease through metastasis monitoring and finding potential mutations for guiding individualized treatment.

Compared with traditional tissue biopsies, liquid biopsy is a relatively non-invasive sample collection method. Liquid biopsy specimens can be obtained with ease and used to monitor diseases in real time dynamically at multiple sampling times (11-13). The most common type of liquid biopsy is circulating tumor DNA (ctDNA), which is a fragmented form of single-stranded or double-stranded DNA that is released into the bloodstream following the necrosis and apoptosis of primary, metastatic or recurrent tumor cells (14,15). ctDNA application is promising in the field of precision oncology due to its potential for use in reflecting the tumor genetic profile and the wide availability of techniques, such as PCR and next-generation sequencing technology (NGS) (16).

However, the accuracy of using ctDNA to reflect all mutational information in the primary tumor lacks direct experimental evidence. Previous studies compared the genetic information between ctDNA and primary or metastatic tumor tissues. Comparison analysis of genetic profiles in prostate, breast and lung cancer showed that the concordance between ctDNA and tissues might depend on alteration types, with considerable concordance in driver DNA alterations but limited concordance in gene amplification and hotspot mutations (17-20). Furthermore, previous analyses suggested that the concordance of hotspot mutations or specific mutations in ctDNA and tumor tissues depends on gene types, for example, 90% concordance of the PIK3CA mutation and 78% concordance of the KRAS mutation were observed in colorectal cancer, and 50% concordance of EGFR mutation was observed in lung cancer (21-24). DNA methylation status and intratumor heterogeneity were also examined in ctDNA. ctDNA could partially reflect the methylation status of a specific gene in breast cancer but had limited value of reflecting intratumor heterogeneity in lung cancer (25,26). These studies revealed that the integrated analysis with both liquid and tumor biopsy might help to obtain a more comprehensive genetic profile of tumors.

ctDNA has exhibited a considerable clinical value as a tumor biomarker in diagnosis and disease monitoring of ovarian cancer (27). However, few studies compared the sequencing results of ctDNA, paired ascites and tumor tissue samples in ovarian cancer (28,29). A parallel analysis of these three types of samples mainly explained the potential of ascites and ctDNA to reflect the mutational landscape in 10 patients (30), although these results require further validation.

Therefore, the present study aimed to detect potential mutant genes in ctDNA samples for clinical application, the data of which were then used to compare against the mutational spectrum in plasma, matching ascites and tumor tissue samples using NGS. The rationale was to further understand the underlying mechanism of the inter-tumor and intra-tumor heterogeneity in ovarian cancer, in addition to its genomic landscape. Overall, the utility of ctDNA to dynamically reflect the overall condition of ovarian cancer and its prospective value in clinical applications, such as metastasis and targeted therapy, were examined.

Materials and methods

Patient enrollment and sample collection

Patients with suspected ovarian cancer were recruited at the Obstetrics and Gynecology Hospital of Fudan University (Shanghai, China) between October 2016 and June 2017. Patients with pathologically confirmed epithelial ovarian cancer were included. Patients with borderline ovarian tumors, malignancies in other organs and uncontrollable infection, and those without consent were excluded. A total of 18 patients were included. In total, 8-10 ml peripheral blood was collected for ctDNA from these 18 patients, ~10 ml matched ascites from 6 of these patients, and 0.5-cm3 tumor tissues from 8 of these patients. The present study was approved by the Ethical Committee of Obstetrics and Gynecology Hospital of Fudan University (approval no. 2016-48; Shanghai, China).

Nucleic acid isolation and quality assessment

Peripheral blood samples were first centrifuged at 800 x g for 10 min at room temperature within 4 h of blood collection to obtain the plasma. The plasma samples were further centrifuged at 16,000 x g for 10 min at room temperature to remove the remaining cellular debris. ctDNA was extracted from the plasma using the QIAamp Circulating Nucleic Acid kit (cat. no. 55114; Qiagen China Co., Ltd.). The quantity and quality of the extracted DNA were assessed using an UV-vis spectrophotometer (Thermo Fisher Scientific, Inc.) and the Agilent 2100 Bioanalyzer.

DNA was extracted from tumor tissue or ascite samples after cell lysis and digestion with the QIAamp DNA Mini Kit (cat. no. 51304; Qiagen China Co., Ltd.) according to the manufacturer's protocols. The quantity and quality of the extracted DNA were assessed as for ctDNA. The extracted DNA from tumor tissues and ascites was sheared using Covaris sonicator (Covaris LLC) with the following settings: 4-8˚C, 10% duty factor, 200 cycles per burst and 360 sec, after further quantification using a Qubit dsDNA HS Assay Kit (cat. no. Q32851; Thermo Fisher Scientific, Inc.). The extracted DNA was stored at -80˚C.

Detection of mutations in DNA sequences using NGS

The SureSelectXT Reagent Kit (cat. no. G9611A; Agilent Technologies, Inc.) was used to construct the NGS libraries according to the SureSelect Target Enrichment workflow instructions. Briefly, after end repair, A-tailing and ligation with adaptors using a SureSelect Library Prep Kit (cat. no. G9684A; Agilent Technologies, Inc.), the DNA was amplified by precapture PCR (1 cycle of 98˚C for 2 min; 10 cycles of 98˚C for 30 sec, 65˚C for 30 sec and 72˚C for 1 min; and then 72˚C for 10 min). The products were then hybridized and captured using the reagents in the aforementioned SureSelectXT Reagent Kit. The captured DNA fractions were amplified by post-capture PCR (1 cycle at 98˚C for 2 min; 16 cycles at 98˚C for 30 sec, 57˚C for 30 sec and 72˚C for 1 min; and then 72˚C for 10 min) using a Herculase II Fusion DNA Polymerase kit (cat. no. 600677; Agilent Technologies, Inc.). Agencourt AMPure XP Beads (cat. no. A63881; Beckman Coulter, Inc.) were used to purify the samples throughout these processes. The libraries were assessed with the DNA 1000 Assay (cat. no. 5067-1504; Agilent Technologies, Inc.) on the Agilent 2100 Bioanalyzer. After quality assessment, the concentration of PCR products was quantified using an Agilent qPCR NGS Library Quantification Kit (cat. no. G4880A; Agilent Technologies, Inc.).

The generated libraries were sequenced with a concentration of 8 pM for 150-bp paired end reads on an Illumina MiSeq sequencer using a MiSeq Reagent Kit (cat. no. MS-102-2002; Illumina, Inc.) according to the manufacturer's recommendations. All samples were sequenced via the OncoGxOne cancer panel by Shanghai Liwen Biotechnology Co., Ltd. After Illumina sequencing, the reads were aligned to the human HG19 reference genome (GRCh37; https://www.ncbi.nlm.nih.gov/grc) using Burrows-Wheeler Aligner v0.7.12 (http://bio-bwa.sourceforge.net). Variant calling was performed using the Genome Analysis Toolkit v3.4 (https://gatk.broadinstitute.org), and was annotated using ANNOVAR (last update, 06/01/17; https://annovar.openbioinformatics.org). The variants with a minor allele frequency <5% were filtered out.

Functional analysis of the mutant genes

Functional analysis of the potential mutant genes was performed using public bioinformatics tools and databases. WebGestalt (2019 version; http://www.webgestalt.org) was used with the following parameters: The functional databases including Gene Ontology (GO), Wikipathway and Kyoto Encyclopedia of Genes and Genomes (KEGG) were selected. The enrichment method was over-representation analysis, where the false discovery rate (FDR) was determined using the Benjamini-Hochberg method. The top 10 categories with FDR <0.05 were identified to be among the enriched categories. The cBioPortal for Cancer Genomics (version 2.0.1; http://www.cbioportal.org/) was used to analyze the gene and drug-target interaction network. The network, which showed the query genes, neighbor genes and drug-target information, was generated by submitting the query genes and adding the drug data in the Network tab (31). ‘metastasis’ and ‘ovarian cancer’ were used as key words in Chilibot (last update 06/18/17; http://www.chilibot.net/) to analyze mutant genes, and the overlap of these two sets was considered as the genes related to ovarian cancer metastasis. Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version 11.0; https://string-db.org) was used to analyze functional interactions with a medium confidence of 0.4. Comparative analysis of the peripheral blood, tumor tissue and ascite samples was performed using Venn diagram (version 2.1; https://bioinfogp.cnb.csic.es/tools/venny/index.html).

Statistical analysis

The data were analyzed using GraphPad Prism 6.0 (GraphPad Software, Inc.) and SPSS 16.0 (SPSS, Inc.). Pearson correlation was used to identify the correlation coefficient and significant difference. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics

The clinicopathological characteristics of the 18 patients with ovarian cancer are summarized in Table I. The mean age of patients was 51.9±11.1 years. Preoperative serum cancer antigen 125 levels were >500 IU/l in 72.2% of the patients. In addition, >80% of the recruited patients were diagnosed with advanced ovarian cancer, at International Federation of Gynecology and Obstetrics stage III or IV. Serous ovarian carcinoma (77.8%) was the most common pathological type, followed by 2 patients with clear cell carcinoma, 1 with mucinous carcinoma and 1 with endometrioid carcinoma. In total, 22.2% of the patients were found with lymph node metastasis.

Table I

Summary of clinicopathological characteristics of patients with ovarian cancer.

Table I

Summary of clinicopathological characteristics of patients with ovarian cancer.

Clinicopathological characteristicsValue
Age range, years24-70
FIGO stage, n (%) 
     II1 (5.6)
     III15 (83.3)
     IV2 (11.1)
Histological type, n (%) 
     Serous carcinoma14 (77.8)
     Clear cell carcinoma2 (11.1)
     Endometrioid carcinoma1 (5.6)
     Mucinous carcinoma1 (5.6)
Lymph nodes metastasis, n (%) 
     Positive4 (22.2)
     Negative4 (22.2)
     Unknown10 (55.6)
Preoperation cancer antigen 125 level, n (%) 
     >1000 IU/l8 (44.4)
     500-1000 IU/l5 (27.8)
     <500 IU/l5 (27.8)

[i] FIGO, International Federation of Gynecology and Obstetrics.

Mutant genes detected in patients with ovarian cancer

A panel of 333 potential oncogenes was used for the targeted NGS of DNA isolated from 18 peripheral blood samples, eight tumor tissue samples and six ascites samples. This panel covers a number of reported genes associated with tumor initiation, those in the clinical investigation phase of cancer drug application, such as AZD6738 against ATR, and those that have been clinically applied for targeted therapy, such as larotrectinib against neurotrophin receptor kinase (Data S1). For example, the small-molecule or antibody drugs targeting MET, SRC, KRAS and BRCA1/2 were approved or in clinical trials. In the present study, for the target regions of the included samples, the coverage reached >99%, whereas the mean sequencing depth was >200X and the alignment rate was >95%.

A total of 104 genes were found to be mutated in the ctDNA samples from ≥1 patient (Fig. 1A). Of these genes, 29 were detected in ≥3 patients, 40 were detected in ≥2 patients, whereas the remaining 64 genes were mutated in only 1 patient. In the ovarian cancer tissue samples, 95 mutant genes were found, among which mutations in 34 genes were found in ≥2 patients (Fig. 1B). A total of 44 mutant genes were found in ascites samples, 23 of which were shared by ≥2 patients (Fig. 1C).

Mutational heterogeneity in patients with ovarian cancer

The mutant genes found in the peripheral blood, tumor tissue and ascites samples were compared to understand the concordance among the mutation spectra and genomic landscapes of the different samples from patients with ovarian cancer. Mutational heterogeneity was widely observed among the specimens and patients. The mutant genes shared by different patients and different sample types within the same individuals are shown in Figs. 1 and 2. The highest similarity was found between the ascites and tumor tissues (51.2%), followed by that between peripheral blood and tumor tissue samples (41.7%). The similarity between peripheral blood and ascite samples was the lowest (39.3%).

In addition, the mutation sites and types in the same gene were different among patients. For example, a missense mutation (D268E) was detected in the PTEN gene in the ctDNA from patients H3 and H14, whilst a deletion mutation (T321fs) in the same gene was detected in the ctDNA from patient H13.

Functional analysis of the mutant genes

To further explore the potential functions of these mutated genes in ovarian cancer samples, the mutant genes found in ≥2 patients were analyzed using the WebGestalt tool. GO analysis revealed that the mutant genes in peripheral blood, tumor tissue and ascites samples mainly participated in the biological processes of ‘Response to stimulus’, ‘biological regulation’, ‘metabolic process’, ‘cell communication’ and ‘cell proliferation’ (Fig. 3A-C). These mutant genes were also analyzed for KEGG and Wikipathway enrichment. The enriched pathways in peripheral blood, tumor tissue and ascites samples were similar and mainly included ‘integrated cancer pathway’, ‘DNA IR-DSBs and cellular response via ATM’, ‘DNA damage response’, ‘Cellular senescence’ and ‘Notch signaling pathway netpath’ (Fig. 3D-F).

Potential of the mutant genes for metastasis evaluation

To find the potential of using mutant genes in the ctDNA samples to monitor metastasis, ‘metastasis’ and ‘ovarian cancer’ were used as key words in Chilibot to analyze the 40 mutant genes presented in ≥2 patients. A cluster covering 10 genes, namely NOTCH2, NOTCH3, lysine methyltransferase 2A (KMT2A), PTEN, androgen receptor (AR), DNA-activated protein kinase catalytic subunit (PRKDC), hepatocyte nuclear factor 1 homeobox A (HNF1A), SRC, insulin receptor substrate 2 (IRS2) and SRY-box transcription factor 10 (SOX10), was obtained. This cluster was then designated as the mutant gene panel for performing correlation analysis between tumor dissemination and metastasis in patients with ovarian cancer. The coverage of this mutant gene panel in the 18 patients who underwent ctDNA sequencing in the present study was 94.4% (17/18). This count was based on tumor lesions >1 cm in the abdominal and pelvic cavities during intraoperative exploration, although miliary lesions were not counted. If the number of tumor lesions was >10 in a patient, then the number of lesions was counted as 10. Pearson correlation analysis found that the number of mutant genes in this mutant gene panel was positively correlated with the number of tumor lesions with a correlation coefficient of 0.851 (P<0.001). This suggests that this panel derived from ctDNA samples may have the potential to be used in reflecting tumor dissemination and metastasis. Furthermore, an interaction network of the mutant genes within this panel was obtained using STRING (Fig. 4A), which indicated the direct or indirect interactions among the mutant genes.

In addition, a validation analysis of this mutant gene panel was performed in matching tumor tissue and ascites samples isolated from the same patient. The coverage of this panel in the paired tumor tissues and ascites was 100%. In total, mutations in six of the genes in the panel, namely NOTCH2, KMT2A, PTEN, AR, PRKDC and SOX10, were found in ascites. By contrast, mutations in eight genes in the panel, specifically NOTCH2, KMT2A, PTEN, AR, PRKDC, HNF1A, IRS2 and SOX10, were found in the ovarian cancer tissues.

Further exploration of mutations associated with targeted therapy

To assess whether the mutant genes had the potential to become targets for ovarian cancer therapy, the cBioPortal online tool was used for exploration. This integrates gene-centric drug-target information. It was found that the genes, except SOX10, in the present panel are either currently under investigation as a target or can already be targeted therapeutically (Fig. 4B). The hormone therapy class of therapeutic agents, such as cyproterone and enzalutamide, can be used for antitumor therapy by targeting AR, whereas there are numerous small-molecule drugs that can target SRC (32,33). NOTCH2 and NOTCH3 can also be targeted for ovarian cancer, since NOTCH3 knockdown has been found to increase the chemosensitivity of paclitaxel-resistant ovarian cancer cells (34,35).

Discussion

Previous studies on the utility of ctDNA of ovarian cancer have mainly focused on early detection and screening (36,37), treatment response evaluation (38-40) and prognosis prediction (41). Higher levels of somatic mutations in ctDNA have been associated with decreased clinical benefit from treatment and shorter progression-free survival or overall survival (40,42-44). By applying NGS or PCR, various types of genetic alterations in ctDNA and/or ovarian cancer tumor tissues have been found, including chromosomal rearrangements (45), chromosomal instability (46), DNA methylation (47,48), and gene mutation and amplification (43,49,50). In the present study, NGS and public database resources based on patient characteristics were used to analyze the profile and function of the mutant genes and mutation patterns in ctDNA, paired ascites and tumor tissue samples from patients with ovarian cancer. By investigating the possible concordance and differences among patients and types of specimens, the mechanism underlying the heterogeneity of ovarian cancer was studied from multiple angles.

ctDNA may have the ability to reveal information regarding the mutation status of all genes involved in the tumor features (51,52). In the present study, ctDNA sequencing was performed in patients with ovarian cancer, where 104 mutant genes were found in all plasma samples. Subsequently, a mutant gene panel that associated the most with tumor metastasis and dissemination was established after analyzing the 40 genes present in ≥2 patient among the ctDNA samples. These results suggest that ctDNA, as a conventional type of non-invasive liquid biopsy, may have the potential for application in monitoring the tumor metastasis process dynamically. However, the dynamic changes in ctDNA expression and mutation profile before and after tumor relapse were not explored in the present study. In addition, the dynamic changes in the ctDNA profile and their association with imaging data and CA125 levels were not analyzed. Therefore, this mutant gene panel requires further validation in future studies.

Through the mutual verification and comparative analysis of sequencing data from ctDNA, ascites and tumor tissues, an understanding of the overall mutational landscape and the heterogeneity of ovarian cancer was at least partially revealed. The heterogeneity of gene mutations in ovarian cancer is commonly observed, where they are manifested as differences in the mutation locations and mutation types (53). In the present study, this heterogeneity was observed from three angles; the inter-tumor heterogeneity among different patients, the intra-tumor heterogeneity among different lesions within the same patient and the clonal heterogeneity in different subclones within the same lesion were all considered. It was then predicted that gene mutations in different types of specimens from the same patient were also heterogeneous in the present study. In particular, some of the gene mutations that could not be detected in tumor tissues could be detected in ctDNA samples, which can be explained by the existence of tumor subclonal heterogeneity and due to the space restriction of tumor sampling. However, it should be noted that the source of ctDNA is not only tumor cells. The cells of the tumor microenvironment, including stromal cells, endothelial cells and immune cells, may release DNA into the circulatory system in patients with cancer (54,55).

ctDNA confers advantages in being able to reflect the overall tumor information compared with the space restrictions of tissue biopsy, particularly in disease monitoring and management (52,56). However, this does not suggest that ctDNA can be used to fully represent all tumor information. Comparative analysis results showed that mutation detection data from ctDNA cannot completely cover the mutant genes found in ascites and tumor tissues. Plasma mutation detection typically has low positive predictive value, meaning that more sensitive ctDNA assays are needed due to the low quantity of ctDNA in a number of patients (57). As a result, parallel sequencing analysis of liquid biopsies, tumor tissues and ascites can be used to unravel the overall tumor information more comprehensively. Combination of clinical, surgical, pathological and molecular features all facilitates the transformation of linear treatments into multidisciplinary and integrated approaches, in turn promoting the advent of individualized therapy to improve the survival outcome (58-60). Therefore, comprehensive analysis into the genetic landscape will likely contribute to the realization of multidisciplinary approaches and personalized medicine in the field of ovarian cancer treatment.

The present study has a number of limitations. A small sample size of matched ctDNA, ascite and tumor tissue samples was evaluated. Low quantities of ascites, insufficient remaining tumor tissues after pathological sampling and quality problems of the tissue samples all contributed to the exclusion of some samples in the present study. As a form of non-invasive examination, peripheral blood samples for ctDNA detection can be easily collected. This maybe one of the reasons why the number of cases with available ctDNA was larger compared with those with paired tumor tissues or ascites. The correlation analysis between mutations in the gene panel in the ctDNA samples and patient survival was lacking in the present study. The cancer panel used in this study included 333 tumor-related genes covering the exons and some introns of coding genes, meaning that mutations in non-coding regions were not explored. This also meant that the ctDNA sequencing data in the present study were not comparable to the sequencing data in TCGA. The sequencing data in TCGA were based on tumor tissues. Therefore, only TCGA was used to assist in screening for mutations due to the difference in subjects between the present study and the external dataset. In addition, since all data in public databases were based on existing reported studies, relying on public databases to assist in screening for genes would run the risk of overlooking significant mutations that were not previously reported.

In conclusion, the present study analyzed the mutant genes of ctDNA and found a molecular panel that can be used to reflect the dissemination and metastasis process of ovarian cancer. In addition, this panel of mutant genes was validated against external data sets, whereas the sequencing data of this panel of mutant genes were compared among ctDNA, matching ascites and tumor tissue samples. A deeper understanding of the overall mutational landscape and heterogeneity was obtained in ovarian cancer. Results from the present study suggest that it is not sufficient to rely solely on the information provided by a specific type of specimens for investigating the genomic landscape and screening for mutations in cancers. Combining the information obtained from multiple types of samples with other diagnostic information facilitates more comprehensive understanding of the pathophysiology of cancer. For future clinical applications of liquid biopsy, a standardized and optimized method for interpreting and utilizing NGS and PCR data is required.

Supplementary Material

Supplementary Data

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by grants from the National Natural Sciences Foundation of China (grant no. 82172747) and the Shanghai Medical Center of Key Programs for Female Reproductive Diseases (grant no. 2017ZZ01016).

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available due to patient privacy protection and the Regulation of the People's Republic of China Administration of Human Genetic Resources (Document Number: Order No. 717 of the State Council of the People's Republic of China) but are available from the corresponding author on reasonable request.

Authors' contributions

CX and XZ designed the study. XJie performed the experiments and analyzed the data. MD and MZ performed data collection and analyzed the data. XJin performed data collection and part of the data analysis. QC performed part of the data analysis. XJie, CX and XZ confirmed the authenticity of the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethical Committee of Obstetrics and Gynecology Hospital of Fudan University (approval no. 2016-48; Shanghai, China). Written informed consent was obtained prior to initiation of the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022.PubMed/NCBI View Article : Google Scholar

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020.PubMed/NCBI View Article : Google Scholar

3 

Jelovac D and Armstrong DK: Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin. 61:183–203. 2011.PubMed/NCBI View Article : Google Scholar

4 

Sundar S, Neal RD and Kehoe S: Diagnosis of ovarian cancer. BMJ. 351(h4443)2015.PubMed/NCBI View Article : Google Scholar

5 

Sabini C, Sorbi F, Cunnea P and Fotopoulou C: Ovarian cancer stem cells: Ready for prime time? Arch Gynecol Obstet. 301:895–899. 2020.PubMed/NCBI View Article : Google Scholar

6 

Lee JY, Yoon JK, Kim B, Kim S, Kim MA, Lim H, Bang D and Song YS: Tumor evolution and intratumor heterogeneity of an epithelial ovarian cancer investigated using next-generation sequencing. BMC Cancer. 15(85)2015.PubMed/NCBI View Article : Google Scholar

7 

Schwarz RF, Ng CK, Cooke SL, Newman S, Temple J, Piskorz AM, Gale D, Sayal K, Murtaza M, Baldwin PJ, et al: Spatial and temporal heterogeneity in high-grade serous ovarian cancer: A phylogenetic analysis. PLoS Med. 12(e1001789)2015.PubMed/NCBI View Article : Google Scholar

8 

Bai H, Li H, Li W, Gui T, Yang J, Cao D and Shen K: The PI3K/AKT/mTOR pathway is a potential predictor of distinct invasive and migratory capacities in human ovarian cancer cell lines. Oncotarget. 6:25520–25532. 2015.PubMed/NCBI View Article : Google Scholar

9 

Luo S, Zhang Y, Yang Y, Zhu S, Liu W, Zhu J, Liang X, Jiang Z, Sun S, Hou X, et al: Clonal tumor mutations in homologous recombination genes predict favorable clinical outcome in ovarian cancer treated with platinum-based chemotherapy. Gynecol Oncol. 158:66–76. 2020.PubMed/NCBI View Article : Google Scholar

10 

Castellarin M, Milne K, Zeng T, Tse K, Mayo M, Zhao Y, Webb JR, Watson PH, Nelson BH and Holt RA: Clonal evolution of high-grade serous ovarian carcinoma from primary to recurrent disease. J Pathol. 229:515–524. 2013.PubMed/NCBI View Article : Google Scholar

11 

Bhardwaj BK, Thankachan S, Venkatesh T and Suresh PS: Liquid biopsy in ovarian cancer. Clin Chim Acta. 510:28–34. 2020.PubMed/NCBI View Article : Google Scholar

12 

Asante DB, Calapre L, Ziman M, Meniawy TM and Gray ES: Liquid biopsy in ovarian cancer using circulating tumor DNA and cells: Ready for prime time? Cancer Lett. 468:59–71. 2020.PubMed/NCBI View Article : Google Scholar

13 

Tuaeva NO, Falzone L, Porozov YB, Nosyrev AE, Trukhan VM, Kovatsi L, Spandidos DA, Drakoulis N, Kalogeraki A, Mamoulakis C, et al: Translational application of circulating DNA in oncology: Review of the last decades achievements. Cells. 8(1251)2019.PubMed/NCBI View Article : Google Scholar

14 

Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ and Yao SL: Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev. 3:67–71. 1994.PubMed/NCBI

15 

Zhou S, Xu B, Qi L, Zhu D, Liu B and Wei J: Next-generation sequencing reveals mutational accordance between cell-free DNA from plasma, malignant pleural effusion and ascites and directs targeted therapy in a gastric cancer patient. Cancer Biol Ther. 20:15–20. 2019.PubMed/NCBI View Article : Google Scholar

16 

Barbosa A, Peixoto A, Pinto P, Pinheiro M and Teixeira MR: Potential clinical applications of circulating cell-free DNA in ovarian cancer patients. Expert Rev Mol Med. 20(e6)2018.PubMed/NCBI View Article : Google Scholar

17 

Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, Foye A, Lloyd P, Nykter M, Beer TM, et al: Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. J Natl Cancer Inst. 109(djx118)2017.PubMed/NCBI View Article : Google Scholar

18 

Xie F, Zhang Y, Mao X, Zheng X, Han-Zhang H, Ye J, Zhao R, Zhang X and Sun J: Comparison of genetic profiles among primary lung tumor, metastatic lymph nodes and circulating tumor DNA in treatment-naïve advanced non-squamous non-small cell lung cancer patients. Lung Cancer. 121:54–60. 2018.PubMed/NCBI View Article : Google Scholar

19 

Xu B, Shan G, Wu Q, Li W, Wang H, Li H, Yang Y, Long Q and Zhao P: Concordance of genomic alterations between circulating tumor DNA and matched tumor tissue in Chinese patients with breast cancer. J Oncol. 2020(4259293)2020.PubMed/NCBI View Article : Google Scholar

20 

Imperial R, Nazer M, Ahmed Z, Kam AE, Pluard TJ, Bahaj W, Levy M, Kuzel TM, Hayden DM, Pappas SG, et al: Matched whole-genome sequencing (WGS) and whole-exome sequencing (WES) of tumor tissue with circulating tumor DNA (ctDNA) analysis: Complementary modalities in clinical practice. Cancers (Basel). 11(1399)2019.PubMed/NCBI View Article : Google Scholar

21 

Wan R, Wang Z, Lee JJ, Wang S, Li Q, Tang F, Wang J, Sun Y, Bai H, Wang D, et al: Comprehensive analysis of the discordance of EGFR mutation status between tumor tissues and matched circulating tumor DNA in advanced non-small cell lung cancer. J Thorac Oncol. 12:1376–1387. 2017.PubMed/NCBI View Article : Google Scholar

22 

Liu HE, Vuppalapaty M, Wilkerson C, Renier C, Chiu M, Lemaire C, Che J, Matsumoto M, Carroll J, Crouse S, et al: Detection of EGFR Mutations in cfDNA and CTCs, and comparison to tumor tissue in non-small-cell-lung-cancer (NSCLC) patients. Front Oncol. 10(572895)2020.PubMed/NCBI View Article : Google Scholar

23 

Kidess-Sigal E, Liu HE, Triboulet MM, Che J, Ramani VC, Visser BC, Poultsides GA, Longacre TA, Marziali A, Vysotskaia V, et al: Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: Comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget. 7:85349–85364. 2016.PubMed/NCBI View Article : Google Scholar

24 

Beije N, Helmijr JC, Weerts MJA, Beaufort CM, Wiggin M, Marziali A, Verhoef C, Sleijfer S, Jansen MPHM and Martens JWM: Somatic mutation detection using various targeted detection assays in paired samples of circulating tumor DNA, primary tumor and metastases from patients undergoing resection of colorectal liver metastases. Mol Oncol. 10:1575–1584. 2016.PubMed/NCBI View Article : Google Scholar

25 

Chimonidou M, Strati A, Malamos N, Kouneli S, Georgoulias V and Lianidou E: Direct comparison study of DNA methylation markers in EpCAM-positive circulating tumour cells, corresponding circulating tumour DNA, and paired primary tumours in breast cancer. Oncotarget. 8:72054–72068. 2017.PubMed/NCBI View Article : Google Scholar

26 

Zhang Y, Chang L, Yang Y, Fang W, Guan Y, Wu A, Hong S, Zhou H, Chen G, Chen X, et al: Intratumor heterogeneity comparison among different subtypes of non-small-cell lung cancer through multi-region tissue and matched ctDNA sequencing. Mol Cancer. 18(7)2019.PubMed/NCBI View Article : Google Scholar

27 

Giannopoulou L and Lianidou ES: Liquid biopsy in ovarian cancer. Adv Clin Chem. 97:13–71. 2020.PubMed/NCBI View Article : Google Scholar

28 

Jagelkova M, Zelinova K, Laucekova Z, Bobrovska M, Dankova Z, Grendar M and Dokus K: Comparison of somatic mutation profiles between formalin-fixed paraffin embedded tissues and plasma cell-free DNA from ovarian cancer patients before and after surgery. Biores Open Access. 9:73–79. 2020.PubMed/NCBI View Article : Google Scholar

29 

Du ZH, Bi FF, Wang L and Yang Q: Next-generation sequencing unravels extensive genetic alteration in recurrent ovarian cancer and unique genetic changes in drug-resistant recurrent ovarian cancer. Mol Genet Genomic Med. 6:638–647. 2018.PubMed/NCBI View Article : Google Scholar

30 

Han MR, Lee SH, Park JY, Hong H, Ho JY, Hur SY and Choi YJ: Clinical implications of circulating tumor DNA from ascites and serial plasma in ovarian cancer. Cancer Res Treat. 52:779–788. 2020.PubMed/NCBI View Article : Google Scholar

31 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6(pl1)2013.PubMed/NCBI View Article : Google Scholar

32 

Mayer EL and Krop IE: Advances in targeting Src in the treatment of breast cancer and other solid malignancies. Clin Cancer Res. 16:3526–3532. 2010.PubMed/NCBI View Article : Google Scholar

33 

Kim LC, Song L and Haura EB: Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol. 6:587–595. 2009.PubMed/NCBI View Article : Google Scholar

34 

Kang H, Jeong JY, Song JY, Kim TH, Kim G, Huh JH, Kwon AY, Jung SG and An HJ: Notch3-specific inhibition using siRNA knockdown or GSI sensitizes paclitaxel-resistant ovarian cancer cells. Mol Carcinog. 55:1196–1209. 2016.PubMed/NCBI View Article : Google Scholar

35 

Zhang J, Yin XJ, Xu CJ, Ning YX, Chen M, Zhang H, Chen SF and Yao LQ: The histone deacetylase SIRT6 inhibits ovarian cancer cell proliferation via down-regulation of Notch 3 expression. Eur Rev Med Pharmaco. 19:818–824. 2015.PubMed/NCBI

36 

Cohen PA, Flowers N, Tong S, Hannan N, Pertile MD and Hui L: Abnormal plasma DNA profiles in early ovarian cancer using a non-invasive prenatal testing platform: Implications for cancer screening. BMC Med. 14(126)2016.PubMed/NCBI View Article : Google Scholar

37 

Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, Anagnostou V, Fiksel J, Cristiano S, Papp E, et al: Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 9(eaan2415)2017.PubMed/NCBI View Article : Google Scholar

38 

Lin KK, Harrell MI, Oza AM, Oaknin A, Ray-Coquard I, Tinker AV, Helman E, Radke MR, Say C, Vo LT, et al: BRCA reversion mutations in circulating tumor DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 9:210–219. 2019.PubMed/NCBI View Article : Google Scholar

39 

Alves MC, Fonseca FLA, Yamada AMTD, Barros LADR, Lopes A, Silva LCFF, Luz AS, Melo Cruz FJS and Del Giglio A: Increased circulating tumor DNA as a noninvasive biomarker of early treatment response in patients with metastatic ovarian carcinoma: A pilot study. Tumour Biol. 42(1010428320919198)2020.PubMed/NCBI View Article : Google Scholar

40 

Kim YM, Lee SW, Lee YJ, Lee HY, Lee JE and Choi EK: Prospective study of the efficacy and utility of TP53 mutations in circulating tumor DNA as a non-invasive biomarker of treatment response monitoring in patients with high-grade serous ovarian carcinoma. J Gynecol Oncol. 30(e32)2019.PubMed/NCBI View Article : Google Scholar

41 

Charo LM, Eskander RN, Okamura R, Patel SP, Nikanjam M, Lanman RB, Piccioni DE, Kato S, McHale MT and Kurzrock R: Clinical implications of plasma circulating tumor DNA in gynecologic cancer patients. Mol Oncol. 15:67–79. 2021.PubMed/NCBI View Article : Google Scholar

42 

Yang F, Tang J, Zhao Z, Zhao C and Xiang Y: Circulating tumor DNA: A noninvasive biomarker for tracking ovarian cancer. Reprod Biol Endocrinol. 19(178)2021.PubMed/NCBI View Article : Google Scholar

43 

Noguchi T, Iwahashi N, Sakai K, Matsuda K, Matsukawa H, Toujima S, Nishio K and Ino K: Comprehensive gene mutation profiling of circulating tumor DNA in ovarian cancer: Its pathological and prognostic impact. Cancers (Basel). 12(3382)2020.PubMed/NCBI View Article : Google Scholar

44 

Salemi R, Falzone L, Madonna G, Polesel J, Cinà D, Mallardo D, Ascierto PA, Libra M and Candido S: MMP-9 as a candidate marker of response to BRAF inhibitors in melanoma patients with BRAFV600E mutation detected in circulating-free DNA. Front Pharmacol. 9(856)2018.PubMed/NCBI View Article : Google Scholar

45 

Harris FR, Kovtun IV, Smadbeck J, Multinu F, Jatoi A, Kosari F, Kalli KR, Murphy SJ, Halling GC, Johnson SH, et al: Quantification of somatic chromosomal rearrangements in circulating cell-free DNA from ovarian cancers. Sci Rep. 6(29831)2016.PubMed/NCBI View Article : Google Scholar

46 

Vanderstichele A, Busschaert P, Smeets D, Landolfo C, Van Nieuwenhuysen E, Leunen K, Neven P, Amant F, Mahner S, Braicu EI, et al: Chromosomal instability in cell-free DNA as a highly specific biomarker for detection of ovarian cancer in women with adnexal masses. Clin Cancer Res. 23:2223–2231. 2017.PubMed/NCBI View Article : Google Scholar

47 

Giannopoulou L, Mastoraki S, Buderath P, Strati A, Pavlakis K, Kasimir-Bauer S and Lianidou ES: ESR1 methylation in primary tumors and paired circulating tumor DNA of patients with high-grade serous ovarian cancer. Gynecol Oncol. 150:355–360. 2018.PubMed/NCBI View Article : Google Scholar

48 

Giannopoulou L, Chebouti I, Pavlakis K, Kasimir-Bauer S and Lianidou ES: RASSF1A promoter methylation in high-grade serous ovarian cancer: A direct comparison study in primary tumors, adjacent morphologically tumor cell-free tissues and paired circulating tumor DNA. Oncotarget. 8:21429–21443. 2017.PubMed/NCBI View Article : Google Scholar

49 

Noguchi T, Sakai K, Iwahashi N, Matsuda K, Matsukawa H, Yahata T, Toujima S, Nishio K and Ino K: Changes in the gene mutation profiles of circulating tumor DNA detected using CAPP-Seq in neoadjuvant chemotherapy-treated advanced ovarian cancer. Oncol Lett. 19:2713–2720. 2020.PubMed/NCBI View Article : Google Scholar

50 

Paracchini L, Beltrame L, Grassi T, Inglesi A, Fruscio R, Landoni F, Ippolito D, Delle Marchette M, Paderno M, Adorni M, et al: Genome-wide copy-number alterations in circulating tumor DNA as a novel biomarker for patients with high-grade serous ovarian cancer. Clin Cancer Res. 27:2549–2559. 2021.PubMed/NCBI View Article : Google Scholar

51 

Sidaway P: Prostate cancer: Mutations in ctDNA reflect features of metastatic disease. Nat Rev Clin Oncol. 14(526)2017.PubMed/NCBI View Article : Google Scholar

52 

ctDNA is a specific and sensitive biomarker in multiple human cancers. Cancer Discov. 4(OF8)2014.PubMed/NCBI View Article : Google Scholar

53 

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, et al: Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 499:214–218. 2013.PubMed/NCBI View Article : Google Scholar

54 

Thierry AR, El Messaoudi S, Gahan PB, Anker P and Stroun M: Origins, structures, and functions of circulating DNA in oncology. Cancer Metast Rev. 35:347–376. 2016.PubMed/NCBI View Article : Google Scholar

55 

Mouliere F and Thierry AR: The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients. Expert Opin Biol Ther. 12 (Suppl 1):S209–S215. 2012.PubMed/NCBI View Article : Google Scholar

56 

ctDNA reveals targetable alterations. Cancer Discov. 10(OF4)2020.PubMed/NCBI View Article : Google Scholar

57 

Franczak C, Filhine-Tressarieu P, Broséus J, Gilson P, Merlin JL and Harlé A: Clinical interest of circulating tumor DNA in oncology. Arch Med Res. 49:297–305. 2018.PubMed/NCBI View Article : Google Scholar

58 

Falzone L, Scandurra G, Lombardo V, Gattuso G, Lavoro A, Distefano AB, Scibilia G and Scollo P: A multidisciplinary approach remains the best strategy to improve and strengthen the management of ovarian cancer (Review). Int J Oncol. 59(53)2021.PubMed/NCBI View Article : Google Scholar

59 

Dondi G, Coluccelli S, De Leo A, Ferrari S, Gruppioni E, Bovicelli A, Godino L, Coadă CA, Morganti AG, Giordano A, et al: An analysis of clinical, surgical, pathological and molecular characteristics of endometrial cancer according to mismatch repair status. A multidisciplinary approach. Int J Mol Sci. 21(7188)2020.PubMed/NCBI View Article : Google Scholar

60 

Heudel PE, Devouassoux-Shisheboran M, Taieb S, Genestie C, Selle F, Morice P, Rouzier R and Ray-Coquard I: Multidisciplinary management of advanced ovarian cancer for an optimal therapeutic strategy. Eur J Gynaecol Oncol. 38:175–180. 2017.PubMed/NCBI

Related Articles

Journal Cover

September-2022
Volume 24 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jie X, Du M, Zhang M, Jin X, Cai Q, Xu C and Zhang X: Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer. Exp Ther Med 24: 542, 2022
APA
Jie, X., Du, M., Zhang, M., Jin, X., Cai, Q., Xu, C., & Zhang, X. (2022). Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer. Experimental and Therapeutic Medicine, 24, 542. https://doi.org/10.3892/etm.2022.11479
MLA
Jie, X., Du, M., Zhang, M., Jin, X., Cai, Q., Xu, C., Zhang, X."Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer". Experimental and Therapeutic Medicine 24.3 (2022): 542.
Chicago
Jie, X., Du, M., Zhang, M., Jin, X., Cai, Q., Xu, C., Zhang, X."Mutation analysis of circulating tumor DNA and paired ascites and tumor tissues in ovarian cancer". Experimental and Therapeutic Medicine 24, no. 3 (2022): 542. https://doi.org/10.3892/etm.2022.11479