Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2021 Apr;11(4):916-932.
doi: 10.1158/2159-8290.CD-20-1559.

Tracking Cancer Evolution through the Disease Course

Affiliations
Review

Tracking Cancer Evolution through the Disease Course

Chris Bailey et al. Cancer Discov. 2021 Apr.

Abstract

During cancer evolution, constituent tumor cells compete under dynamic selection pressures. Phenotypic variation can be observed as intratumor heterogeneity, which is propagated by genome instability leading to mutations, somatic copy-number alterations, and epigenomic changes. TRACERx was set up in 2014 to observe the relationship between intratumor heterogeneity and patient outcome. By integrating multiregion sequencing of primary tumors with longitudinal sampling of a prospectively recruited patient cohort, cancer evolution can be tracked from early- to late-stage disease and through therapy. Here we review some of the key features of the studies and look to the future of the field. SIGNIFICANCE: Cancers evolve and adapt to environmental challenges such as immune surveillance and treatment pressures. The TRACERx studies track cancer evolution in a clinical setting, through primary disease to recurrence. Through multiregion and longitudinal sampling, evolutionary processes have been detailed in the tumor and the immune microenvironment in non-small cell lung cancer and clear-cell renal cell carcinoma. TRACERx has revealed the potential therapeutic utility of targeting clonal neoantigens and ctDNA detection in the adjuvant setting as a minimal residual disease detection tool primed for translation into clinical trials.

PubMed Disclaimer

Conflict of interest statement

Conflicts of Interests

CS is Royal Society Napier Research Professor. This work was supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (FC001169), the UK Medical Research Council (FC001169), and the Wellcome Trust (FC001169). CS is funded by Cancer Research UK (TRACERx, PEACE and CRUK Cancer Immunotherapy Catalyst Network), Cancer Research UK Lung Cancer Centre of Excellence, the Rosetrees Trust, Butterfield and Stoneygate Trusts, NovoNordisk Foundation (ID16584), Royal Society Professorship Enhancement Award (RP/EA/180007), the NIHR BRC at University College London Hospitals, and the CRUK-UCL Centre, Experimental Cancer Medicine Centre, and the Breast Cancer Research Foundation (BCRF). This research is supported by a Stand Up To Cancer‐LUNGevity-American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT23-17). Stand Up To Cancer is a division of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the Scientific Partner of SU2C. CS receives funding from the European Research Council (ERC) under the European Union’s Seventh Framework Programme (FP7/2007-2013) Consolidator Grant (FP7-THESEUS-617844), European Commission ITN (FP7-PloidyNet 607722), an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 835297), and Chromavision from the European Union’s Horizon 2020 research and innovation programme (grant agreement 665233). CS acknowledges grant support from Pfizer, AstraZeneca, Bristol Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Archer Dx Inc (collaboration in minimal residual disease sequencing technologies) and Ono Pharmaceutical, is an AstraZeneca Advisory Board member and Chief Investigator for the MeRmaiD1 clinical trial, has consulted for Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol Myers Squibb, Celgene, AstraZeneca, Illumina, Amgen, Genentech, Roche-Ventana, GRAIL, Medicxi, Bicycle Therapeutics, and the Sarah Cannon Research Institute, has stock options in Apogen Biotechnologies, Epic Bioscience, GRAIL, and has stock options and is co-founder of Achilles Therapeutics. CS holds European patents relating to assay technology to detect tumour recurrence (PCT/GB2017/053289); to targeting neoantigens (PCT/EP2016/059401), identifying patent response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), identifying patients who respond to cancer treatment (PCT/GB2018/051912), a US patent relating to detecting tumour mutations (PCT/US2017/28013) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892).

ST is funded by Cancer Research UK (grant reference number C50947/A18176), the National Institute for Health Research (NIHR) Biomedical Research Centre at the Royal Marsden Hospital and Institute of Cancer Research (grant reference number A109), the Kidney and Melanoma Cancer Fund of The Royal Marsden Cancer Charity, The Rosetrees Trust (grant reference number A2204), Ventana Medical Systems Inc (grant reference numbers 10467 and 10530), the National Institute of Health U01 Award and Melanoma Research Alliance. ST has received speaking fees from Roche, Astra Zeneca, Novartis and Ipsen. ST has the following patents filed: Indel mutations as a therapeutic target and predictive biomarker PCTGB2018/051892 and PCTGB2018/051893 and Clear Cell Renal Cell Carcinoma Biomarkers P113326GB.

MJH has received funding from Cancer Research UK, National Institute for Health Research, Rosetrees Trust, UKI NETs and NIHR University College London Hospitals Biomedical Research Centre. MJH is a member of the Scientific Advisory Board and Steering Committee for Achilles Therapeutics.

Figures

Figure 1
Figure 1. Outline of TRACERx Lung and PEACE autopsy studies.
1. Following a diagnosis of early stage non-small cell lung cancer, patients undergo surgery to resect their primary tumour (with adjuvant chemotherapy, depending on disease stage). Multiple regions from the primary tumour are sampled. In the event of recurrent disease, biopsies are obtained where possible. Patients that are enrolled in the PEACE study have multiple sites of disease sampled for analysis. Tumour samples are processed for whole exome sequencing, RNA-seq and tissue microarrays. 2. Recruitment sites for TRACERx and PEACE in the UK. 3. The core analysis of tumour samples involves calling clonal and subclonal mutation and copy number alterations, with construction of phylogenetic trees. Combining tissue microarray, RNA-seq data and other techniques has facilitated the study of immune and spatial heterogeneity. PV-CTC, pulmonary vein circulating tumour cells.
Figure 2
Figure 2. Tumour evolution in non-small cell lung cancer.
Evolutionary processes in non-small cell lung cancer are outlined. Top, subclonal dynamics over time can be represented by a fish plot, however a single sample in time only provides a snapshot. From this snapshot tumour phylogeny can be inferred. Bottom, evolutionary processes generating immune and genomic heterogeneity are described as part of a ‘tree’. Events that occur in the ‘trunk’ are clonal; i.e. they occur within every cell in the tumour. Through tumour evolution, subclones can emerge through selection; events that occur in these subclones are known as ‘branch’ events.
Figure 3
Figure 3. Clinical trials in tumour evolution.
Outlined are key trials that have been associated with the TRACERx project. Top, the MERMAID-1 trial incorporates bespoke ctDNA panels as MRD detection, following on from the work by Abbosh et al (52). Left, the THETIS and CHIRON trials are first-in-man studies that look at the efficacy of autologous clonal neoantigen reactive T cells in non-small cell lung cancer and melanoma. Clonal neoantigens are identified following tumour sequencing. T cells are subsequently expanded with neo-antigenic peptides and returned to the patient. Right, the DARWIN-II trial investigates the impact of intra-tumour heterogeneity in targeted therapy response in relapsed non small cell lung cancer, combining data from primary tumours sampled in the TRACERx Lung project.

Similar articles

  • Tracking the Evolution of Non-Small-Cell Lung Cancer.
    Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, Shafi S, Johnson DH, Mitter R, Rosenthal R, Salm M, Horswell S, Escudero M, Matthews N, Rowan A, Chambers T, Moore DA, Turajlic S, Xu H, Lee SM, Forster MD, Ahmad T, Hiley CT, Abbosh C, Falzon M, Borg E, Marafioti T, Lawrence D, Hayward M, Kolvekar S, Panagiotopoulos N, Janes SM, Thakrar R, Ahmed A, Blackhall F, Summers Y, Shah R, Joseph L, Quinn AM, Crosbie PA, Naidu B, Middleton G, Langman G, Trotter S, Nicolson M, Remmen H, Kerr K, Chetty M, Gomersall L, Fennell DA, Nakas A, Rathinam S, Anand G, Khan S, Russell P, Ezhil V, Ismail B, Irvin-Sellers M, Prakash V, Lester JF, Kornaszewska M, Attanoos R, Adams H, Davies H, Dentro S, Taniere P, O'Sullivan B, Lowe HL, Hartley JA, Iles N, Bell H, Ngai Y, Shaw JA, Herrero J, Szallasi Z, Schwarz RF, Stewart A, Quezada SA, Le Quesne J, Van Loo P, Dive C, Hackshaw A, Swanton C; TRACERx Consortium. Jamal-Hanjani M, et al. N Engl J Med. 2017 Jun 1;376(22):2109-2121. doi: 10.1056/NEJMoa1616288. Epub 2017 Apr 26. N Engl J Med. 2017. PMID: 28445112
  • Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution.
    Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, Le Quesne J, Moore DA, Veeriah S, Rosenthal R, Marafioti T, Kirkizlar E, Watkins TBK, McGranahan N, Ward S, Martinson L, Riley J, Fraioli F, Al Bakir M, Grönroos E, Zambrana F, Endozo R, Bi WL, Fennessy FM, Sponer N, Johnson D, Laycock J, Shafi S, Czyzewska-Khan J, Rowan A, Chambers T, Matthews N, Turajlic S, Hiley C, Lee SM, Forster MD, Ahmad T, Falzon M, Borg E, Lawrence D, Hayward M, Kolvekar S, Panagiotopoulos N, Janes SM, Thakrar R, Ahmed A, Blackhall F, Summers Y, Hafez D, Naik A, Ganguly A, Kareht S, Shah R, Joseph L, Marie Quinn A, Crosbie PA, Naidu B, Middleton G, Langman G, Trotter S, Nicolson M, Remmen H, Kerr K, Chetty M, Gomersall L, Fennell DA, Nakas A, Rathinam S, Anand G, Khan S, Russell P, Ezhil V, Ismail B, Irvin-Sellers M, Prakash V, Lester JF, Kornaszewska M, Attanoos R, Adams H, Davies H, Oukrif D, Akarca AU, Hartley JA, Lowe HL, Lock S, Iles N, Bell H, Ngai Y, Elgar G, Szallasi Z, Schwarz RF, Herrero J, Stewart A, Quezada SA, Peggs KS, Van Loo P, Dive C, Lin CJ, Rabinowitz M, Aerts HJWL, Hackshaw A, Shaw JA, Zimmermann BG; TRACERx consortium; PEACE consortium; Swanton C. Abbosh C, et al. Nature. 2017 Apr 26;545(7655):446-451. doi: 10.1038/nature22364. Nature. 2017. PMID: 28445469 Free PMC article.
  • Tracking genomic cancer evolution for precision medicine: the lung TRACERx study.
    Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, Middleton G, de Bruin E, Le Quesne J, Shafi S, Falzon M, Horswell S, Blackhall F, Khan I, Janes S, Nicolson M, Lawrence D, Forster M, Fennell D, Lee SM, Lester J, Kerr K, Muller S, Iles N, Smith S, Murugaesu N, Mitter R, Salm M, Stuart A, Matthews N, Adams H, Ahmad T, Attanoos R, Bennett J, Birkbak NJ, Booton R, Brady G, Buchan K, Capitano A, Chetty M, Cobbold M, Crosbie P, Davies H, Denison A, Djearman M, Goldman J, Haswell T, Joseph L, Kornaszewska M, Krebs M, Langman G, MacKenzie M, Millar J, Morgan B, Naidu B, Nonaka D, Peggs K, Pritchard C, Remmen H, Rowan A, Shah R, Smith E, Summers Y, Taylor M, Veeriah S, Waller D, Wilcox B, Wilcox M, Woolhouse I, McGranahan N, Swanton C. Jamal-Hanjani M, et al. PLoS Biol. 2014 Jul 8;12(7):e1001906. doi: 10.1371/journal.pbio.1001906. eCollection 2014 Jul. PLoS Biol. 2014. PMID: 25003521 Free PMC article.
  • Variability in the Solid Cancer Cell Population and Its Consequences for Cancer Diagnostics and Treatment.
    Brychtová V, Valík V, Vojtěšek B. Brychtová V, et al. Klin Onkol. 2018 Winter;31(Suppl 2):5-13. doi: 10.14735/amko20182S5. Klin Onkol. 2018. PMID: 31023018 Review. English.
  • Translational implications of tumor heterogeneity.
    Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C. Jamal-Hanjani M, et al. Clin Cancer Res. 2015 Mar 15;21(6):1258-66. doi: 10.1158/1078-0432.CCR-14-1429. Clin Cancer Res. 2015. PMID: 25770293 Free PMC article. Review.

Cited by

  • Decoding the interplay between genetic and non-genetic drivers of metastasis.
    Karras P, Black JRM, McGranahan N, Marine JC. Karras P, et al. Nature. 2024 May;629(8012):543-554. doi: 10.1038/s41586-024-07302-6. Epub 2024 May 15. Nature. 2024. PMID: 38750233 Review.
  • Ecological and evolutionary dynamics to design and improve ovarian cancer treatment.
    Han GYQ, Alexander M, Gattozzi J, Day M, Kirsch E, Tafreshi N, Chalar R, Rahni S, Gossner G, Burke W, Damaghi M. Han GYQ, et al. Clin Transl Med. 2024 Sep;14(9):e70012. doi: 10.1002/ctm2.70012. Clin Transl Med. 2024. PMID: 39210542 Free PMC article. Review.
  • Benchmarking copy number aberrations inference tools using single-cell multi-omics datasets.
    Song M, Ma S, Wang G, Wang Y, Yang Z, Xie B, Guo T, Huang X, Zhang L. Song M, et al. Brief Bioinform. 2025 Mar 4;26(2):bbaf076. doi: 10.1093/bib/bbaf076. Brief Bioinform. 2025. PMID: 40037644 Free PMC article.
  • ACT-Discover: identifying karyotype heterogeneity in pancreatic cancer evolution using ctDNA.
    Huebner A, Black JRM, Sarno F, Pazo R, Juez I, Medina L, Garcia-Carbonero R, Guillén C, Feliú J, Alonso C, Arenillas C, Moreno-Cárdenas AB, Verdaguer H, Macarulla T, Hidalgo M, McGranahan N, Toledo RA. Huebner A, et al. Genome Med. 2023 Apr 20;15(1):27. doi: 10.1186/s13073-023-01171-w. Genome Med. 2023. PMID: 37081523 Free PMC article.
  • The evolution of non-small cell lung cancer metastases in TRACERx.
    Al Bakir M, Huebner A, Martínez-Ruiz C, Grigoriadis K, Watkins TBK, Pich O, Moore DA, Veeriah S, Ward S, Laycock J, Johnson D, Rowan A, Razaq M, Akther M, Naceur-Lombardelli C, Prymas P, Toncheva A, Hessey S, Dietzen M, Colliver E, Frankell AM, Bunkum A, Lim EL, Karasaki T, Abbosh C, Hiley CT, Hill MS, Cook DE, Wilson GA, Salgado R, Nye E, Stone RK, Fennell DA, Price G, Kerr KM, Naidu B, Middleton G, Summers Y, Lindsay CR, Blackhall FH, Cave J, Blyth KG, Nair A, Ahmed A, Taylor MN, Procter AJ, Falzon M, Lawrence D, Navani N, Thakrar RM, Janes SM, Papadatos-Pastos D, Forster MD, Lee SM, Ahmad T, Quezada SA, Peggs KS, Van Loo P, Dive C, Hackshaw A, Birkbak NJ, Zaccaria S; TRACERx Consortium; Jamal-Hanjani M, McGranahan N, Swanton C. Al Bakir M, et al. Nature. 2023 Apr;616(7957):534-542. doi: 10.1038/s41586-023-05729-x. Epub 2023 Apr 12. Nature. 2023. PMID: 37046095 Free PMC article.

References

    1. Turajlic S, Sottoriva A, Graham T, Swanton C. Resolving genetic heterogeneity in cancer. Nat Rev Genet. 2019;20:404–16. [Internet]. Available from: http://www.nature.com/articles/s41576-019-0114-6. - PubMed
    1. Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, et al. Tracking Genomic Cancer Evolution for Precision Medicine: The Lung TRACERx Study. PLoS Biol. 2014;12:e1001906. doi: 10.1371/journal.pbio.1001906. [Internet] - DOI - PMC - PubMed
    1. Turajlic S, Xu H, Litchfield K, Rowan A, Horswell S, Chambers T, et al. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal. Cell. 2018;173:595–610.:e11. [Internet]. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0092867418303751. - PMC - PubMed
    1. McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, Mastrogianakis GM, et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–8. [Internet]. Available from: http://www.nature.com/articles/nature07385. - PMC - PubMed
    1. Campbell PJ, Getz G, Korbel JO, Stuart JM, Jennings JL, Stein LD, et al. Pan-cancer analysis of whole genomes. Nature. 2020;578:82–93. [Internet]. Available from: http://www.nature.com/articles/s41586-020-1969-6. - PMC - PubMed