Skip to content
Licensed Unlicensed Requires Authentication Published online by De Gruyter April 5, 2024

Exploring evolutionary trajectories in ovarian cancer patients by longitudinal analysis of ctDNA

  • Oliver Kutz , Stephan Drukewitz , Alexander Krüger , Daniela Aust , Doreen William , Sandra Oster , Evelin Schröck , Gustavo Baretton , Theresa Link , Pauline Wimberger and Jan Dominik Kuhlmann EMAIL logo

Abstract

Objectives

We analysed whether temporal heterogeneity of ctDNA encodes evolutionary patterns in ovarian cancer.

Methods

Targeted sequencing of 275 cancer-associated genes was performed in a primary tumor biopsy and in ctDNA of six longitudinal plasma samples from 15 patients, using the Illumina platform.

Results

While there was low overall concordance between the mutational spectrum of the primary tumor biopsies vs. ctDNA, TP53 variants were the most commonly shared somatic alterations. Up to three variant clusters were detected in each tumor biopsy, likely representing predominant clones of the primary tumor, most of them harbouring a TP53 variant. By tracing these clusters in ctDNA, we propose that liquid biopsy may allow to assess the contribution of ancestral clones of the tumor to relapsed abdominal masses, revealing two evolutionary patterns. In pattern#1, clusters detected in the primary tumor biopsy were likely relapse seeding clones, as they contributed a major share to ctDNA at relapse. In pattern#2, similar clusters were present in tumors and ctDNA; however, they were entirely cleared from liquid biopsy after chemotherapy and were undetectable at relapse. ctDNA private variants were present among both patterns, with some of them mirroring subclonal expansions after chemotherapy.

Conclusions

We demonstrate that tracing the temporal heterogeneity of ctDNA, even below exome scale resolution, deciphers evolutionary trajectories in ovarian cancer. Furthermore, we describe two evolutionary patterns that may help to identify relapse seeding clones for targeted therapy.


Corresponding author: Prof. Dr. rer. nat. Jan Dominik Kuhlmann, Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr., 74, 01307 Dresden, Germany; National Center for Tumour Diseases (NCT), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany; German Cancer Consortium (DKTK) Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany, Phone: +49 – 351 458 2434, Fax: +49 – 351 458 5844, E-mail:

Funding source: Deutsche Krebshilfe

Award Identifier / Grant number: 70113616, 70113636

  1. Research ethics: The study was approved by the Local Research Ethics Committee in Dresden, Germany (EK74032013, EK23602012) and was performed in accordance with good clinical practice guidelines, national laws, and the Declaration of Helsinki.

  2. Informed consent: Written informed consent was obtained from all patients.

  3. Author contributions: JDK, OK and SD and DA made substantial contributions to the conception and design of the study. OK, JDK, AK, SO contributed to the experimental work. OK, AK, JDK, DA, SD contributed to the acquisition of data. JDK, OK, DW, SD analyzed and interpreted the results. JDK, OK, TL, PW, GB, ES were involved in drafting the manuscript, creating figures or revising the manuscript. All authors read and approved the manuscript in its final version.

  4. Competing interests: The authors state no conflict of interest.

  5. Research funding: The study was supported by Deutsche Krebshilfe (reference numbers: 70113616, 70113636).

  6. Data availability: The sequence raw data of this study have been deposited in the SRA (Sequence Read Archive) with the submission number SUB13842319.

References

1. Siegel, R, Ma, J, Zou, Z, Jemal, A. Cancer statistics, 2014. CA Cancer J Clin 2014;64:9–29. https://doi.org/10.3322/caac.21208.Search in Google Scholar PubMed

2. Ray-Coquard, I, Pautier, P, Pignata, S, Perol, D, Gonzalez-Martin, A, Berger, R, et al.. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med 2019;381:2416–28. https://doi.org/10.1056/nejmoa1911361.Search in Google Scholar

3. Moore, K, Colombo, N, Scambia, G, Kim, BG, Oaknin, A, Friedlander, M, et al.. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018;379:2495–505. https://doi.org/10.1056/nejmoa1810858.Search in Google Scholar PubMed

4. Gonzalez-Martin, A, Desauw, C, Heitz, F, Cropet, C, Gargiulo, P, Berger, R, et al.. Maintenance olaparib plus bevacizumab in patients with newly diagnosed advanced high-grade ovarian cancer: main analysis of second progression-free survival in the phase III PAOLA-1/ENGOT-ov25 trial. Eur J Cancer 2022;174:221–31. https://doi.org/10.1016/j.ejca.2022.07.022.Search in Google Scholar PubMed

5. Perren, TJ, Swart, AM, Pfisterer, J, Ledermann, JA, Pujade-Lauraine, E, Kristensen, G, et al.. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011;365:2484–96. https://doi.org/10.1056/nejmoa1103799.Search in Google Scholar PubMed

6. Ray-Coquard, I, Leary, A, Pignata, S, Cropet, C, Gonzalez-Martin, A, Marth, C, et al.. Olaparib plus bevacizumab first-line maintenance in ovarian cancer: final overall survival results from the PAOLA-1/ENGOT-ov25 trial. Ann Oncol 2023;34:681–92. https://doi.org/10.1016/j.annonc.2023.05.005.Search in Google Scholar PubMed

7. Bashashati, A, Ha, G, Tone, A, Ding, J, Prentice, LM, Roth, A, et al.. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J Pathol 2013;231:21–34. https://doi.org/10.1002/path.4230.Search in Google Scholar PubMed PubMed Central

8. Ahmed, AA, Etemadmoghadam, D, Temple, J, Lynch, AG, Riad, M, Sharma, R, et al.. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010;221:49–56. https://doi.org/10.1002/path.2696.Search in Google Scholar PubMed PubMed Central

9. Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474:609–15. https://doi.org/10.1038/nature10166.Search in Google Scholar PubMed PubMed Central

10. Jacobs, IJ, Kohler, MF, Wiseman, RW, Marks, JR, Whitaker, R, Kerns, BA, et al.. Clonal origin of epithelial ovarian carcinoma: analysis by loss of heterozygosity, p53 mutation, and X-chromosome inactivation. J Natl Cancer Inst 1992;84:1793–8. https://doi.org/10.1093/jnci/84.23.1793.Search in Google Scholar PubMed

11. Khalique, L, Ayhan, A, Whittaker, JC, Singh, N, Jacobs, IJ, Gayther, SA, et al.. The clonal evolution of metastases from primary serous epithelial ovarian cancers. Int J Cancer 2009;124:1579–86. https://doi.org/10.1002/ijc.24148.Search in Google Scholar PubMed

12. McPherson, A, Roth, A, Laks, E, Masud, T, Bashashati, A, Zhang, AW, et al.. Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer. Nat Genet 2016;48:758–67. https://doi.org/10.1038/ng.3573.Search in Google Scholar PubMed

13. Burrell, RA, Swanton, C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol Oncol 2014;8:1095–111. https://doi.org/10.1016/j.molonc.2014.06.005.Search in Google Scholar PubMed PubMed Central

14. Al Bakir, M, Huebner, A, Martinez-Ruiz, C, Grigoriadis, K, Watkins, TBK, Pich, O, et al.. The evolution of non-small cell lung cancer metastases in TRACERx. Nature 2023;616:534–42. https://doi.org/10.1038/s41586-023-05729-x.Search in Google Scholar PubMed PubMed Central

15. Frankell, AM, Dietzen, M, Al Bakir, M, Lim, EL, Karasaki, T, Ward, S, et al.. The evolution of lung cancer and impact of subclonal selection in TRACERx. Nature 2023;616:525–33. https://doi.org/10.1038/s41586-023-05783-5.Search in Google Scholar PubMed PubMed Central

16. McGranahan, N, Favero, F, de Bruin, EC, Birkbak, NJ, Szallasi, Z, Swanton, C. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci Transl Med 2015;7:283ra54. https://doi.org/10.1126/scitranslmed.aaa1408.Search in Google Scholar PubMed PubMed Central

17. Lohr, JG, Stojanov, P, Carter, SL, Cruz-Gordillo, P, Lawrence, MS, Auclair, D, et al.. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell 2014;25:91–101. https://doi.org/10.1016/j.ccr.2013.12.015.Search in Google Scholar PubMed PubMed Central

18. Achimas-Cadariu, P, Kubelac, P, Irimie, A, Berindan-Neagoe, I, Ruhli, F. Evolutionary perspectives, heterogeneity and ovarian cancer: a complicated tale from past to present. J Ovarian Res 2022;15:67. https://doi.org/10.1186/s13048-022-01004-1.Search in Google Scholar PubMed PubMed Central

19. Lahtinen, A, Lavikka, K, Virtanen, A, Li, Y, Jamalzadeh, S, Skorda, A, et al.. Evolutionary states and trajectories characterized by distinct pathways stratify patients with ovarian high grade serous carcinoma. Cancer Cell 2023;41:1103–17 e12. https://doi.org/10.1016/j.ccell.2023.04.017.Search in Google Scholar PubMed

20. Paracchini, L, Mannarino, L, Beltrame, L, Landoni, F, Fruscio, R, Grassi, T, et al.. Targeted mutational analysis of circulating tumor DNA to decipher temporal heterogeneity of high-grade serous ovarian cancer. Cancers 2022;14:3697. https://doi.org/10.3390/cancers14153697.Search in Google Scholar PubMed PubMed Central

21. Sabatier, R, Garnier, S, Guille, A, Carbuccia, N, Pakradouni, J, Adelaide, J, et al.. Whole-genome/exome analysis of circulating tumor DNA and comparison to tumor genomics from patients with heavily pre-treated ovarian cancer: subset analysis of the PERMED-01 trial. Front Oncol 2022;12:946257. https://doi.org/10.3389/fonc.2022.946257.Search in Google Scholar PubMed PubMed Central

22. Mithraprabhu, S, Sirdesai, S, Chen, M, Khong, T, Spencer, A. Circulating tumour DNA analysis for tumour genome characterisation and monitoring disease burden in extramedullary multiple myeloma. Int J Mol Sci 2018;19:1858. https://doi.org/10.3390/ijms19071858.Search in Google Scholar PubMed PubMed Central

23. Herberts, C, Annala, M, Sipola, J, Ng, SWS, Chen, XE, Nurminen, A, et al.. Deep whole-genome ctDNA chronology of treatment-resistant prostate cancer. Nature 2022;608:199–208. https://doi.org/10.1038/s41586-022-04975-9.Search in Google Scholar PubMed

24. Cresswell, GD, Nichol, D, Spiteri, I, Tari, H, Zapata, L, Heide, T, et al.. Mapping the breast cancer metastatic cascade onto ctDNA using genetic and epigenetic clonal tracking. Nat Commun 2020;11:1446. https://doi.org/10.1038/s41467-020-15047-9.Search in Google Scholar PubMed PubMed Central

25. FIGO Committee on Gynecologic Oncology. Current FIGO staging for cancer of the vagina, fallopian tube, ovary, and gestational trophoblastic neoplasia. Int J Gynaecol Obstet 2009;105:3–4. https://doi.org/10.1016/j.ijgo.2008.12.015.Search in Google Scholar PubMed

26. Prat, J, Oncology FCoG. Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet 2014;124:1–5. https://doi.org/10.1016/j.ijgo.2013.10.001.Search in Google Scholar PubMed

27. Smith, T, Heger, A, Sudbery, I. UMI-tools: modeling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy. Genome Res 2017;27:491–9. https://doi.org/10.1101/gr.209601.116.Search in Google Scholar PubMed PubMed Central

28. Bolger, AM, Lohse, M, Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20. https://doi.org/10.1093/bioinformatics/btu170.Search in Google Scholar PubMed PubMed Central

29. Li, H, Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009;25:1754–60. https://doi.org/10.1093/bioinformatics/btp324.Search in Google Scholar PubMed PubMed Central

30. McKenna, A, Hanna, M, Banks, E, Sivachenko, A, Cibulskis, K, Kernytsky, A, et al.. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. https://doi.org/10.1101/gr.107524.110.Search in Google Scholar PubMed PubMed Central

31. Koboldt, DC, Zhang, Q, Larson, DE, Shen, D, McLellan, MD, Lin, L, et al.. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 2012;22:568–76. https://doi.org/10.1101/gr.129684.111.Search in Google Scholar PubMed PubMed Central

32. Li, H, Handsaker, B, Wysoker, A, Fennell, T, Ruan, J, Homer, N, et al.. The sequence alignment/map format and SAMtools. Bioinformatics 2009;25:2078–9. https://doi.org/10.1093/bioinformatics/btp352.Search in Google Scholar PubMed PubMed Central

33. Genomes Project, C, Auton, A, Brooks, LD, Durbin, RM, Garrison, EP, Kang, HM, et al.. A global reference for human genetic variation. Nature 2015;526:68–74. https://doi.org/10.1038/nature15393.Search in Google Scholar PubMed PubMed Central

34. Landrum, MJ, Chitipiralla, S, Brown, GR, Chen, C, Gu, B, Hart, J, et al.. ClinVar: improvements to accessing data. Nucleic Acids Res 2020;48:D835–44. https://doi.org/10.1093/nar/gkz972.Search in Google Scholar PubMed PubMed Central

35. Forbes, SA, Beare, D, Boutselakis, H, Bamford, S, Bindal, N, Tate, J, et al.. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 2017;45:D777–83. https://doi.org/10.1093/nar/gkw1121.Search in Google Scholar PubMed PubMed Central

36. Karczewski, KJ, Francioli, LC, Tiao, G, Cummings, BB, Alfoldi, J, Wang, Q, et al.. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 2020;581:434–43. https://doi.org/10.1038/s41586-020-2308-7.Search in Google Scholar PubMed PubMed Central

37. Cingolani, P, Platts, A, Wang le, L, Coon, M, Nguyen, T, Wang, L, et al.. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly 2012;6:80–92. https://doi.org/10.4161/fly.19695.Search in Google Scholar PubMed PubMed Central

38. Crowdis, J, He, MX, Reardon, B, Van Allen, EM. CoMut: visualizing integrated molecular information with comutation plots. Bioinformatics 2020;36:4348–9. https://doi.org/10.1093/bioinformatics/btaa554.Search in Google Scholar PubMed PubMed Central

39. Miller, CA, White, BS, Dees, ND, Griffith, M, Welch, JS, Griffith, OL, et al.. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput Biol 2014;10:e1003665. https://doi.org/10.1371/journal.pcbi.1003665.Search in Google Scholar PubMed PubMed Central

40. Miller, CA, McMichael, J, Dang, HX, Maher, CA, Ding, L, Ley, TJ, et al.. Visualizing tumor evolution with the fishplot package for R. BMC Genom 2016;17:880. https://doi.org/10.1186/s12864-016-3195-z.Search in Google Scholar PubMed PubMed Central

41. Abraham, A, Pedregosa, F, Eickenberg, M, Gervais, P, Mueller, A, Kossaifi, J, et al.. Machine learning for neuroimaging with scikit-learn. Front Neuroinform 2014;8:14. https://doi.org/10.3389/fninf.2014.00014.Search in Google Scholar PubMed PubMed Central

42. Ruark, E, Snape, K, Humburg, P, Loveday, C, Bajrami, I, Brough, R, et al.. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature 2013;493:406–10. https://doi.org/10.1038/nature11725.Search in Google Scholar PubMed PubMed Central

43. Castellarin, M, Milne, K, Zeng, T, Tse, K, Mayo, M, Zhao, Y, et al.. Clonal evolution of high-grade serous ovarian carcinoma from primary to recurrent disease. J Pathol 2013;229:515–24. https://doi.org/10.1002/path.4105.Search in Google Scholar PubMed

44. Zill, OA, Banks, KC, Fairclough, SR, Mortimer, SA, Vowles, JV, Mokhtari, R, et al.. The landscape of actionable genomic alterations in cell-free circulating tumor DNA from 21,807 advanced cancer patients. Clin Cancer Res 2018;24:3528–38. https://doi.org/10.1158/1078-0432.ccr-17-3837.Search in Google Scholar

45. Woodcock, DJ, Riabchenko, E, Taavitsainen, S, Kankainen, M, Gundem, G, Brewer, DS, et al.. Prostate cancer evolution from multilineage primary to single lineage metastases with implications for liquid biopsy. Nat Commun 2020;11:5070. https://doi.org/10.1038/s41467-020-18843-5.Search in Google Scholar PubMed PubMed Central

46. Marchi, G, Rajavuori, A, Nguyen, MTN, Huhtinen, K, Oksa, S, Hietanen, S, et al.. Extensive mutational ctDNA profiles reflect High-grade serous cancer tumors and reveal emerging mutations at recurrence. Transl Oncol 2024;39:101814. https://doi.org/10.1016/j.tranon.2023.101814.Search in Google Scholar PubMed PubMed Central

47. Cooke, SL, Ng, CK, Melnyk, N, Garcia, MJ, Hardcastle, T, Temple, J, et al.. Genomic analysis of genetic heterogeneity and evolution in high-grade serous ovarian carcinoma. Oncogene 2010;29:4905–13. https://doi.org/10.1038/onc.2010.245.Search in Google Scholar PubMed PubMed Central

48. Huang, R, Zhu, L, Zhang, Y. XIST lost induces ovarian cancer stem cells to acquire taxol resistance via a KMT2C-dependent way. Cancer Cell Int 2020;20:436. https://doi.org/10.1186/s12935-020-01500-8.Search in Google Scholar PubMed PubMed Central

49. Wimberger, P, Wehling, M, Lehmann, N, Kimmig, R, Schmalfeldt, B, Burges, A, et al.. Influence of residual tumor on outcome in ovarian cancer patients with FIGO stage IV disease: an exploratory analysis of the AGO-OVAR (Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group). Ann Surg Oncol 2010;17:1642–8. https://doi.org/10.1245/s10434-010-0964-9.Search in Google Scholar PubMed


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/cclm-2023-1266).


Received: 2023-11-09
Accepted: 2024-03-12
Published Online: 2024-04-05

© 2024 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 30.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2023-1266/html
Scroll to top button