Skip to main content

Advertisement

Log in

Significance of Distinct Liquid Biopsy Compartments in Evaluating Somatic Mutations for Targeted Therapy Selection in Cancer of Unknown Primary

  • Original Research
  • Published:
Journal of Gastrointestinal Cancer Aims and scope Submit manuscript

Abstract

Purpose

Cancer of unknown primary (CUP) accounts for 2–5% of all cancer diagnoses, wherein standard investigations fail to reveal the original tumor site. Basket trials allocate targeted therapeutics based on actionable somatic mutations, independent of tumor entity. These trials, however, mostly rely on variants identified in tissue biopsies. Since liquid biopsies (LB) represent the overall tumor genomic landscape, they may provide an ideal diagnostic source in CUP patients. To identify the most informative liquid biopsy compartment, we compared the utility of genomic variant analysis for therapy stratification in two LB compartments (circulating cell-free (cf) and extracellular vesicle (ev) DNA).

Methods

CfDNA and evDNA from 23 CUP patients were analyzed using a targeted gene panel covering 151 genes. Identified genetic variants were interpreted regarding diagnostic and therapeutic relevance using the MetaKB knowledgebase.

Results

LB revealed a total of 22 somatic mutations in evDNA and/or cfDNA in 11/23 patients. Out of the 22 identified somatic variants, 14 are classified as Tier I druggable somatic variants. Comparison of variants identified in evDNA and cfDNA revealed an overlap of 58% of somatic variants in both LB compartments, whereas over 40% of variants were only found in one or the other compartment.

Conclusion

We observed substantial overlap between somatic variants identified in evDNA and cfDNA of CUP patients. Nonetheless, interrogation of both LB compartments can potentially increase the rate of druggable alterations, stressing the significance of liquid biopsies for possible primary-independent basket and umbrella trial inclusion.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Data Availability

The datasets generated and analyzed during the current study are available from the corresponding authors on reasonable request.

Abbreviations

AMP:

Association for Molecular Pathology

ASCO:

American Society of Clinical Oncology

CAP:

College of American Pathologists

cfDNA:

Circulating cell-free DNA

CNV:

Copy number variation

CUP:

Cancer of unknown primary

evDNA:

Extracellular vesicle DNA

NGS:

Next-generation sequencing

PBMC:

Peripheral blood mononuclear cells

SNV:

Single-nucleotide variant

References

  1. Binder C, Matthes KL, Korol D, Rohrmann S, Moch H. Cancer of unknown primary-Epidemiological trends and relevance of comprehensive genomic profiling. Cancer Med. 2018;7:4814–24.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Pavlidis N, Pentheroudakis G. Cancer of unknown primary site. Lancet. 2012;379:1428–35.

    Article  PubMed  Google Scholar 

  3. Varadhachary GR, Raber MN. Carcinoma of unknown primary site. N Engl J Med. 2014;371:2040.

    Article  PubMed  Google Scholar 

  4. Blaszyk H, Hartmann A, Bjornsson J. Cancer of unknown primary: clinicopathologic correlations. APMIS. 2003;111:1089–94.

    Article  PubMed  Google Scholar 

  5. Pentheroudakis G, Golfinopoulos V, Pavlidis N. Switching benchmarks in cancer of unknown primary: from autopsy to microarray. Eur J Cancer. 2007;43:2026–36.

    Article  PubMed  Google Scholar 

  6. Hemminki K, Riihimaki M, Sundquist K, Hemminki A. Site-specific survival rates for cancer of unknown primary according to location of metastases. Int J Cancer. 2013;133:182–9.

    Article  CAS  PubMed  Google Scholar 

  7. Raghav K, et al. Cancer of unknown primary in adolescents and young adults: clinicopathological features, prognostic factors and survival outcomes. PLoS ONE. 2016;11: e0154985.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Fizazi K, et al. Cancers of unknown primary site: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v133–8.

    Article  PubMed  Google Scholar 

  9. Greco FA, Lennington WJ, Spigel DR, Hainsworth JD. Poorly differentiated neoplasms of unknown primary site: diagnostic usefulness of a molecular cancer classifier assay. Mol Diagn Ther. 2015;19:91–7.

    Article  CAS  PubMed  Google Scholar 

  10. Hainsworth JD, Fizazi K. Treatment for patients with unknown primary cancer and favorable prognostic factors. Semin Oncol. 2009;36:44–51.

    Article  CAS  PubMed  Google Scholar 

  11. Culine S. Prognostic factors in unknown primary cancer. Semin Oncol. 2009;36:60–4.

    Article  CAS  PubMed  Google Scholar 

  12. Losa F, et al. SEOM clinical guideline on unknown primary cancer (2017). Clin Transl Oncol. 2018;20:89.

    Article  CAS  PubMed  Google Scholar 

  13. Greco FA, Lennington WJ, Spigel DR, Hainsworth JD. Molecular profiling diagnosis in unknown primary cancer: accuracy and ability to complement standard pathology. J Natl Cancer Inst. 2013;105:782–90.

    Article  CAS  PubMed  Google Scholar 

  14. Varadhachary GR, et al. Molecular profiling of carcinoma of unknown primary and correlation with clinical evaluation. J Clin Oncol. 2008;26:4442–8.

    Article  CAS  PubMed  Google Scholar 

  15. Murtaza M, et al. Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer. Nat Commun. 2015;6.

  16. Pennec SL, et al. Intratumor heterogeneity and clonal evolution in an aggressive papillary thyroid cancer and matched metastases. Endocr Relat Cancer. 2015;22:205–16.

    Article  PubMed  Google Scholar 

  17. Chaudhuri AA, et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017;7:1394–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dawson SJ, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199–209.

    Article  CAS  PubMed  Google Scholar 

  19. Tie J, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med. 2016;8:346ra92.

  20. Forshew T, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4.

  21. Fackler MJ, et al. Novel methylated biomarkers and a robust assay to detect circulating tumor DNA in metastatic breast cancer. Cancer Res. 2014;74:2160–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. San Lucas FA, et al. Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes. Ann Oncol. 2016;27:635–641.

  23. Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J Transl Med. 2011;9:86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kahlert C. Liquid biopsy: is there an advantage to analyzing circulating exosomal DNA compared to cfDNA or are they the same? Cancer Res. 2019;79:2462–5.

    Article  CAS  PubMed  Google Scholar 

  25. Cohen JD, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–930.

  26. Bernard V, et al. Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterol. 2019;156:108–118 e4.

  27. Wagner AH, et al. A harmonized meta-knowledgebase of clinical interpretations of somatic genomic variants in cancer. Nat Genet. 2020;52:448–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Castillo J, et al. Surfaceome profiling enables isolation of cancer-specific exosomal cargo in liquid biopsies from pancreatic cancer patients. Ann Oncol Off J Eur Soc Med Oncol. 2018;29:223–9.

    Article  CAS  Google Scholar 

  29. Semaan A, et al. Defining the comprehensive genomic landscapes of pancreatic ductal adenocarcinoma using real-world endoscopic aspiration samples. Clin Cancer Res. 2021;27:1082–93.

    Article  PubMed  Google Scholar 

  30. Li MM, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer. J Mol Diagnostics. 2017;19:4–23.

    Article  CAS  Google Scholar 

  31. Zehir A, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yang H, Ye D, Guan KL, Xiong Y. IDH1 and IDH2 mutations in tumorigenesis: mechanistic insights and clinical perspectives. Clin Cancer Res. 2012;18:5562.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jiao Y, et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet. 2013;45:1470–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zou S, et al. Mutational landscape of intrahepatic cholangiocarcinoma. Nat Commun. 2014;(51)5:1–11.

  35. Sallman DA, et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J Clin Oncol. 2021;39:1584–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lindemann A, et al. COTI-2, a novel thiosemicarbazone derivative, exhibits antitumor activity in HNSCC through p53-dependent and -independent mechanisms. Clin Cancer Res. 2019;25:5650–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chapman PB, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. 2011;364:2507–2516. https://doi.org/10.1056/NEJMoa1103782.

  38. Solomon BJ, et al. First-line crizotinib versus chemotherapy in ALK -positive lung cancer. N Engl J Med. 2014;371:2167–77.

    Article  PubMed  Google Scholar 

  39. Hainsworth JD, et al. Molecular gene expression profiling to predict the tissue of origin and direct site-specific therapy in patients with carcinoma of unknown primary site: a prospective trial of the Sarah Cannon research institute. J Clin Oncol. 2013;31:217–23.

    Article  CAS  PubMed  Google Scholar 

  40. Yoon HH, et al. Gene expression profiling identifies responsive patients with cancer of unknown primary treated with carboplatin, paclitaxel, and everolimus: NCCTG N0871 (alliance). Ann Oncol. 2016;27:339–44.

    Article  CAS  PubMed  Google Scholar 

  41. Hayashi H, et al. Randomized phase II trial comparing site-specific treatment based on gene expression profiling with carboplatin and paclitaxel for patients with cancer of unknown primary site. J Clin Oncol. 2019;37:570–9.

    Article  CAS  PubMed  Google Scholar 

  42. Fizazi K, et al. A phase III trial of empiric chemotherapy with cisplatin and gemcitabine or systemic treatment tailored by molecular gene expression analysis in patients with carcinomas of an unknown primary (CUP) site (GEFCAPI 04). 2019. https://doi.org/10.1093/annonc/mdz394.

  43. Woodcock J, LaVange LM. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377:62–70.

    Article  CAS  PubMed  Google Scholar 

  44. Pauli C, et al. A challenging task: identifying patients with cancer of unknown primary (CUP) according to ESMO guidelines: the CUPISCO trial experience. Oncologist. 2021;26: e769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wolfers J, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7:297–303.

    Article  CAS  PubMed  Google Scholar 

  46. Kato S, et al. Utility of genomic analysis in circulating tumor DNA from patients with carcinoma of unknown primary. Cancer Res. 2017;77:4238–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nguyen L, Van Hoeck A, Cuppen E. Machine learning-based tissue of origin classification for cancer of unknown primary diagnostics using genome-wide mutation features. Nat Commun. 2022;13:1–12.

    Article  CAS  Google Scholar 

  48. Laprovitera N, et al. Genetic characterization of cancer of unknown primary using liquid biopsy approaches. Front Cell Dev Biol. 2021;9.

Download references

Acknowledgements

We thank our patients and their families. We dedicate this study to the memory of our beloved colleague Dr. Gauri R. Varadhachary who was responsible for initiating this study and was always there for her patients.

Funding

A.M. is supported by the MD Anderson Pancreatic Cancer Moon Shot Program, the Sheikh Khalifa bin Zayed Al-Nahyan Foundation, and the NIH (R01CA218230, U01CA196403, U01CA200468, and P50CA221707). V. Bernard is supported by the NIH (U54CA096300, U54CA096297, and T32CA217789). J.J.L. is supported by the NIH (T32CA009599). A.S. is supported by the German Research Foundation (SE-2616/2–1).

Author information

Authors and Affiliations

Authors

Contributions

F.R.K. performed experiments and data analysis and wrote the manuscript, B.M.S. performed experiments, V. Branchi conducted data analysis, V. Bernard and J.J.L. edited the manuscript. K.P.S.R. helped with clinical annotations, A.M. provided funding and scientific guidance and edited the manuscript. A.S. and P.A.G, conceptualized and guided the work and edited the manuscript. All authors reviewed and approved the final manuscript.

Corresponding authors

Correspondence to Paola A. Guerrero or Alexander Semaan.

Ethics declarations

Competing Interests

A.M. receives royalties for a pancreatic cancer biomarker test from Cosmos Wisdom Biotechnology, and this financial relationship is managed and monitored by the UTMDACC Conflict of Interest Committee. A.M. is also listed as an inventor on a patent that has been licensed by Johns Hopkins University to Thrive Earlier Detection (an Exact Sciences Company). A.M. is a consultant for Freenome and Tezcat. All other authors declare no conflicts of interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Significance

Analysis of genomic variants in liquid biopsies has potential as a diagnostic tool for CUP samples. We compared cfDNA and evDNA by a targeted NGS approach and identified druggable targets that could help to stratify patients for inclusion into clinical trials.

Supplementary Information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kolbinger, F.R., Bernard, V., Lee, J.J. et al. Significance of Distinct Liquid Biopsy Compartments in Evaluating Somatic Mutations for Targeted Therapy Selection in Cancer of Unknown Primary. J Gastrointest Canc 54, 1276–1285 (2023). https://doi.org/10.1007/s12029-023-00922-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12029-023-00922-7

Keywords

Navigation