Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer

Abstract

The incidence of rectal cancer is increasing in patients younger than 50 years. Locally advanced rectal cancer is still treated with neoadjuvant radiation, chemotherapy and surgery, but recent evidence suggests that patients with a complete response can avoid surgery permanently. To define correlates of response to neoadjuvant therapy, we analyzed genomic and transcriptomic profiles of 738 untreated rectal cancers. APC mutations were less frequent in the lower than in the middle and upper rectum, which could explain the more aggressive behavior of distal tumors. No somatic alterations had significant associations with response to neoadjuvant therapy in a treatment-agnostic manner, but KRAS mutations were associated with faster relapse in patients treated with neoadjuvant chemoradiation followed by consolidative chemotherapy. Overexpression of IGF2 and L1CAM was associated with decreased response to neoadjuvant therapy. RNA-sequencing estimates of immune infiltration identified a subset of microsatellite-stable immune hot tumors with increased response and prolonged disease-free survival.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The genomic landscape of rectal cancer.
Fig. 2: Differences in WNT signaling across the rectum.
Fig. 3: Clinical and genomic determinants of response to neoadjuvant therapy in LARC.
Fig. 4: Transcriptomic determinants of response to neoadjuvant therapy in LARC.
Fig. 5: Immune profiling identifies a subset of immune hot pMMR/MSS LARC tumors with favorable outcomes from neoadjuvant therapy.

Similar content being viewed by others

Data availability

All genomic results and associated clinical data for all of the patients in this study have been deposited in the cBioPortal for Cancer Genomics55,56 and are publicly available for browsing and bulk download at https://www.cbioportal.org/study/summary?id=rectal_msk_2022. The raw RNA sequencing data have also been deposited in GEO (accession number GSE209746 available at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE209746). The raw DNA sequencing data are protected; de-identified data are available under restricted access to protect patient privacy in accordance with Federal and State law. These data can be requested for research use from the corresponding author. Data will be shared for a span of 2 years within 2 weeks of execution of a data transfer agreement with MSK, which will retain all title and rights to the data and results from their use. The OncoKB knowledge base that we used to annotate genomic alterations is publicly available at https://www.oncokb.org/. TCGA data used for comparison are available via the Genomic Data Commons Portal (https://portal.gdc.cancer.gov/).

Code availability

The mutational signature decomposition code can be found at https://github.com/mskcc/tempoSig. The OncoKB annotator tool is also available through its own GitHub repository at https://github.com/oncokb. Additional custom written tools and programs used for the analysis of MSK-IMPACT data are available through the MSK GitHub repository at https://github.com/mskcc.

References

  1. Siegel, R. L., Miller, K. D., Fuchs, H. E. & Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 71, 7–33 (2021).

    Article  PubMed  Google Scholar 

  2. Saad El Din, K. et al. Trends in the epidemiology of young-onset colorectal cancer: a worldwide systematic review. BMC Cancer 20, 288 (2020).

    Article  PubMed  Google Scholar 

  3. Marr, R. et al. The modern abdominoperineal excision: the next challenge after total mesorectal excision. Ann. Surg. 242, 74–82 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Smith, J. J. et al. Assessment of a watch-and-wait strategy for rectal cancer in patients with a complete response after neoadjuvant therapy. JAMA Oncol. 5, e185896 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Garcia-Aguilar, J. et al. Organ preservation in patients with rectal adenocarcinoma treated with total neoadjuvant therapy. J. Clin. Oncol. https://doi.org/10.1200/JCO.22.00032 (2022).

  6. Garcia-Aguilar, J. et al. Effect of adding mFOLFOX6 after neoadjuvant chemoradiation in locally advanced rectal cancer: a multicentre, phase 2 trial. Lancet Oncol. 16, 957–966 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Probst, C. P. et al. Extended intervals after neoadjuvant therapy in locally advanced rectal cancer: the key to improved tumor response and potential organ preservation. J. Am. Coll. Surg. 221, 430–440 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Garcia-Aguilar, J. et al. Identification of a biomarker profile associated with resistance to neoadjuvant chemoradiation therapy in rectal cancer. Ann. Surg. 254, 486–492 (2011).

    Article  PubMed  Google Scholar 

  9. Chow, O. S. et al. KRAS and combined KRAS/TP53 mutations in locally advanced rectal cancer are independently associated with decreased response to neoadjuvant therapy. Ann. Surg. Oncol. 23, 2548–2555 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kamran, S. C. et al. Integrative molecular characterization of resistance to neoadjuvant chemoradiation in rectal cancer. Clin. Cancer Res. 25, 5561–5571 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

    Article  CAS  Google Scholar 

  12. Liu, Y. et al. Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell 33, 721–735 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Garcia-Aguilar, J. et al. Organ preservation for clinical T2N0 distal rectal cancer using neoadjuvant chemoradiotherapy and local excision (ACOSOG Z6041): results of an open-label, single-arm, multi-institutional, phase 2 trial. Lancet Oncol. 16, 1537–1546 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 173, 321–337. (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mondaca, S. et al. Specific mutations in APC, but not alterations in DNA damage response, associate with outcomes of patients with metastatic colorectal cancer. Gastroenterology 159, 1975–1978 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Bielski, C. M. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 50, 1189–1195 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689. (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chakravarty, D. et al. Oncokb: a precision oncology knowledge base. JCO Precis. Oncol. 2017, PO.17.00011 (2017).

    Google Scholar 

  21. Infante, J. R. et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol. 13, 773–781 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Cleary, J. M. et al. Differential outcomes in codon 12/13 and codon 61 NRAS-mutated cancers in the phase II NCI-MATCH trial of binimetinib in patients with NRAS-mutated tumors. Clin. Cancer Res. 27, 2996–3004 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bendell, J. C. et al. Phase I, dose-escalation study of BKM120, an oral pan-class I PI3K inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 30, 282–290 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. van Geel, R. M. J. M. et al. A phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov. 7, 610–619 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Yaeger, R. et al. Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell 33, 125–136 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Roxburgh, C. S. D. et al. Changes in the multidisciplinary management of rectal cancer from 2009 to 2015 and associated improvements in short-term outcomes. Colorectal Dis. 21, 1140–1150 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cercek, A. et al. Mismatch repair-deficient rectal cancer and resistance to neoadjuvant chemotherapy. Clin. Cancer Res. 26, 3271–3279 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Guinney, J. et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350–1356 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Korkeila, E. et al. Expression of carbonic anhydrase IX suggests poor outcome in rectal cancer. Br. J. Cancer 100, 874–880 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee-Kong, S. A. et al. Hypoxia-related proteins in patients with rectal cancer undergoing neoadjuvant combined modality therapy. Dis. Colon Rectum 55, 990–995 (2012).

    Article  PubMed  Google Scholar 

  31. Ganesh, K. et al. L1CAM defines the regenerative origin of metastasis-initiating cells in colorectal cancer. Nat. Cancer 1, 28–45 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bindea, G. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer–immunity cycle. Immunity 39, 1–10 (2013).

    Article  PubMed  CAS  Google Scholar 

  35. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nishimura, T. et al. The critical role of Th1-dominant immunity in tumor immunology. Cancer Chemother. Pharmacol. 46, S52–S61 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Lee, M. S., Menter, D. G. & Kopetz, S. Right versus left colon cancer biology: integrating the consensus molecular subtypes. J. Natl Compr. Canc. Netw. 15, 411–419 (2017).

    Article  PubMed  Google Scholar 

  38. Νikolouzakis, Τ. Κ. et al. Detailed and applied anatomy for improved rectal cancer treatment. Ann. Gastroenterol. 32, 431–440 (2019).

    Google Scholar 

  39. Cheng, L.-J. et al. Distinct prognosis of high versus mid/low rectal cancer: a propensity score-matched cohort study. J. Gastrointest. Surg. 23, 1474–1484 (2019).

    Article  PubMed  Google Scholar 

  40. Yang, H. et al. Influence of tumor location on short- and long-term outcomes after laparoscopic surgery for rectal cancer: a propensity score matched cohort study. BMC Cancer 20, 761 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Nagtegaal, I. D. et al. Low rectal cancer: a call for a change of approach in abdominoperineal resection. J. Clin. Oncol. 23, 9257–9264 (2005).

    Article  PubMed  Google Scholar 

  42. Patel, S. V. et al. Distance to the anal verge is associated with pathologic complete response to neoadjuvant therapy in locally advanced rectal cancer. J. Surg. Oncol. 114, 637–641 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim, J. K. et al. KRAS mutant rectal cancer cells interact with surrounding fibroblasts to deplete the extracellular matrix. Mol. Oncol. 15, 2766–2781 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Francois, Y. et al. Influence of the interval between preoperative radiation therapy and surgery on downstaging and on the rate of sphincter-sparing surgery for rectal cancer: the Lyon R90-01 randomized trial. J. Clin. Oncol. 17, 2396 (1999).

    Article  CAS  PubMed  Google Scholar 

  45. Moore, H. G. et al. Rate of pathologic complete response with increased interval between preoperative combined modality therapy and rectal cancer resection. Dis. Colon Rectum 47, 279–286 (2004).

    Article  PubMed  Google Scholar 

  46. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ganesh, K. et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 16, 361–375 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Rahma, O. E. et al. Use of total neoadjuvant therapy for locally advanced rectal cancer: initial results from the pembrolizumab arm of a phase 2 randomized clinical trial. JAMA Oncol. 7, 1225–1230 (2021).

    Article  PubMed  Google Scholar 

  49. Pagès, F. et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).

    Article  PubMed  Google Scholar 

  50. El Sissy, C. et al. A diagnostic biopsy-adapted immunoscore predicts response to neoadjuvant treatment and selects patients with rectal cancer eligible for a watch-and-wait strategy. Clin. Cancer Res. 26, 5198–5207 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Kirilovsky, A. et al. The “Immunoscore” in rectal cancer: could we search quality beyond quantity of life? Oncotarget 13, 18–31 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zanella, E. R. et al. IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti-EGFR therapies. Sci. Transl. Med. 7, 272ra12 (2015).

    Article  PubMed  CAS  Google Scholar 

  53. Smith, J. J. et al. Organ Preservation in Rectal Adenocarcinoma: a phase II randomized controlled trial evaluating 3-year disease-free survival in patients with locally advanced rectal cancer treated with chemoradiation plus induction or consolidation chemotherapy, and total mesorectal excision or nonoperative management. BMC Cancer 15, 767 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Schrag, D. et al. Challenges and solutions in the design and execution of the PROSPECT phase II/III neoadjuvant rectal cancer trial (NCCTG N1048/Alliance). Clin. Trials 16, 165–175 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Cerami, E. et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  56. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Cheng, D. T. et al. Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Bielski, C. M. et al. Widespread selection for oncogenic mutant allele imbalance in cancer. Cancer Cell 34, 852–862 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Niu, B. et al. MSIsensor: microsatellite instability detection using paired tumor–normal sequence data. Bioinformatics 30, 1015–1016 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Middha, S. et al. Reliable pan-cancer microsatellite instability assessment by using targeted next-generation sequencing data. JCO Precis. Oncol. 2017, PO.17.00084 (2017).

    Google Scholar 

  63. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Eide, P. W., Bruun, J., Lothe, R. A. & Sveen, A. CMScaller: an R package for consensus molecular subtyping of colorectal cancer pre-clinical models. Sci. Rep. 7, 16618 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Salgado, R. et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann. Oncol. 26, 259–271 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Budinska, E. et al. Gene expression patterns unveil a new level of molecular heterogeneity in colorectal cancer. J. Pathol. 231, 63–76 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the use of services provided by the Molecular Cytology Core Facility, funded by the National Cancer Institute (NCI) Cancer Center Support Grant (CCSG, P30 CA008748-53). The authors also acknowledge the use of the Integrated Genomics Operation Core, funded by the NCI Cancer Center Support Grant (CCSG, P30 CA08748), Cycle for Survival, and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. W.K.C. is supported by a National Institutes of Health (NIH) research training grant (T32 GM132083). P.B.R. is supported by an NIH/NCI early career development award (K08 CA255574). J.J.S. is supported by an NIH/NCI R37 248289 award. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Author information

Authors and Affiliations

Authors

Contributions

W.K.C., F.S.-V. and J.G.-A. conceived and designed the study in concert with N.S., R.Y. and J.J.S. A.C., M.R.W., M.W., I.H.W., E.P.P., G.M.N., P.B.P., Q.S., K.G., R.Y., J.J.S. and J.G.-A. provided patient samples and clinical information. C.-T.C., S.-H.C., M.R.M., J.K.K., O.S.C. and M.F.B. generated genomic data. J.K.K., M.R.M., C.-T.C., D.M.O., K.I.G. and J.G.-A. annotated patient samples and curated the clinical data. W.K.C., F.S.-V., H.W., K.G., X.Q., F.W., P.B.R. and J.G.-A. performed data analysis and interpretation. J.S. and E.V. carried out the histopathologic analyses. D.N.K. interpreted the immunology results. S.D.S. carried out the project management. W.K.C., H.W., F.W., X.Q., R.P., R.K., M.T.A.D., Y.-J.H., A.L., M.F.B., F.S.V. and N.S., provided bioinformatic support for data analysis, annotation, visualization and dissemination. M.F.B., D.B.S. and N.S. provided support as part of the institutional MSK-IMPACT sequencing initiative. W.K.C., H.W., R.Y., J.J.S., N.S., F.S.-V. and J.G.-A. wrote the manuscript. All authors reviewed and approved the manuscript. N.S., R.Y., F.S.-V. and J.G.-A. provided resources and funding for this work. F.S.-V. and J.G.-A. supervised the study.

Corresponding authors

Correspondence to Francisco Sanchez-Vega or Julio Garcia-Aguilar.

Ethics declarations

Competing interests

D.N.K. has been a consultant for Merck Sharp & Dohme with regard to intellectual property rights and for AbbVie and PsiOxus Therapeutics Ltd with regard to provision of services. E.P.P. has received support from Intuitive Surgical. Q.S. reports a consulting or advisory role with Yiviva, Boehringer Ingelheim Pharmaceuticals, Regeneron Pharmaceuticals, Hoosier Cancer Research Network (to self), an honorarium or speaker role with Chugai Pharmaceutical Co., stocks from Johnson & Johnson, Amgen and Merck & CO. (to self), and research funds from Celgene/BMS, Roche/Genentech, Janssen and Novartis (to institution). D.B.S. has consulted for and received honoraria from Pfizer, Lilly/Loxo Oncology, Vividion Therapeutics, Scorpion Therapeutics and BridgeBio. M.F.B. has consulted for Eli Lilly and PetDx, and has received research funding from Grail not related to the work presented. P.B.R. is an EMD Serono consultant and reports support for travel from Elekta and Philips healthcare and prior research funding from EMD Serono. R.Y. has been an advisor for Pfizer, Mirati Therapeutics and Natera, and has received research support from Pfizer, Boehringer Ingelheim and Forte Biosciences. J.J.S. has received travel support from Intuitive Surgical for fellow education and has served as a clinical advisor for Guardant Health. J.G.-A. has received an honorarium for being a consultant with Medtronics, Ethicon, Johnson & Johnson and Intuitive Surgical, and owns stock in Intuitive Surgical. All other authors have no competing interests.

Peer review

Peer review information

Nature Medicine thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Anna Maria Ranzoni, in collaboration with the Nature Medicine team.

Additional information

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

Extended data

Extended Data Fig. 1 Cohort overview and patient breakdown by analyses.

((a) Overview of the different sample sets used for the different analyses described in the manuscript, including sample sizes and reasons for exclusion. (b) Venn diagrams showing overlaps for patients with available MSK-IMPACT, WES, RNA-Seq and neoadjuvant therapy (NAT) outcome data. Color bars show the distribution of different relevant clinical variables. (c-h) Same as B, but restricted to the subset of patients used in specific analyses described in the manuscript. Thick red contours drawn on top of the Venn diagrams are used to highlight the set of patients used in each case. The G# in the titles refer to the columns found in Supplementary Table 1.

Extended Data Fig. 2 Additional insights into the genomic landscape of rectal cancer.

(a) Overview of driver alterations in rectal cancer stratified by tumor stage. (b) Distribution of clonal versus driver mutations for the most frequently mutated genes in our rectal cancer cohort. (c) Fraction of samples with two driver mutations in selected genes where both are clonal, both are subclonal or only one is clonal. (d) Distribution of KRAS mutations stratified by affected codon and specific amino acid change. Blue vertical bars show the fraction of clonal versus subclonal mutations. Red and gray bars show the fraction of samples with allelic imbalance (mutant selection). (e) Distribution of mutational signatures for samples in the WES cohort. Samples were ordered from left to right in terms of decreasing SBS1 signature (mitotic clock) and stratified according to dMMR/MSI status.

Extended Data Fig. 3 Comparison of colon and rectal adenocarcinomas.

(a) Clinicopathological features for right colon, left colon, and rectum samples. (b) Differences in first site of metastasis stratified by primary tumor location. (c) Tumor mutational burden (TMB) and FGA in pMMR/MSS tumors from the right colon (n = 121), left colon (n = 187), and rectum (n = 449). Statistical significance was assessed using a two-sided Mann–Whitney U-test. (d) Frequency of somatic alterations in oncogenic signaling pathways by anatomic location. Significant results were denoted as * indicating q < 0.05, ** indicating q < 0.01, *** indicating q < 0.005, and **** indicating q < 0.001. (e) Frequency of RAS/RAF alterations in hypermutated and non-hypermutated tumors stratified by tumor location. (f) Copy number profiles for tumors in the analyzed cohorts. (g) Frequency of copy number alterations affecting the p and q arms of chromosome 20 by anatomic location. (h) FGA as a function of TP53 status, stratified by missense versus truncating and mono-allelic versus biallelic inactivation, for tumors from the right colon (wild-type n = 39, missense n = 8, missense biallelic n = 33, truncating n = 1, truncating biallelic n = 17), left colon (wild-type n = 32, missense n = 10, missense biallelic n = 77, truncating n = 5, truncating biallelic n = 29) and rectum (wild-type n = 73, missense n = 44, missense biallelic n = 175, truncating n = 12, truncating biallelic n = 81). (i) Fraction of dMMR/MSI tumors by rectal segment. (j) Distance to the anal verge by APC status in the validation cohort of metastatic patients. APC WT (n = 43) were compared to APC altered (n = 115) using a two-sided Mann–Whitney U-test, * indicates p = 0.0029. (k) Distribution of APC mutations by genomic location in tumors from the right colon, left colon, upper rectum, middle rectum, and lower rectum. In panels (B), (D) and (G), statistical significance was assessed using a two-sided Fisher’s exact test and p values were corrected for multiple testing using false discovery rate. In panels (C), (H) and (J), boxplots’ center lines indicate medians, edges indicate the interquartile range, and whiskers extend to the highest and lowest values not considered outliers.

Extended Data Fig. 4 Clinical and genomic determinants of response to NAT in LARC.

(a) Frequency of somatic alterations in rectal cancer driver genes for the patients used in our analyses of clinical outcomes, stratified by cohort. (b) Frequency of somatic alterations in oncogenic signaling pathways for the patients used in our analyses of clinical outcomes, stratified by cohort. (c) Left panel shows results from a multivariate analysis of associations between CR and a combination of clinicopathological and genomic features using a logistic regression model. The error bars indicate the 95% confidence interval. Right panel shows results from a multivariate analysis of associations between DFS and a combination of clinicopathological and genomic features using a Cox proportional hazards model. The results shown in this panel were obtained using patients treated with CRT-CNCT. (d) The left panel shows a multivariate analysis of associations between CR and a combination of clinicopathological and genomic features using a logistic regression model. The error bars indicate the 95% confidence interval. The right panel shows results from a multivariate analysis of associations between DFS and a combination of clinicopathological and genomic features using a Cox proportional hazards model. The results shown in this panel were obtained using patients treated with INCT-CRT.

Extended Data Fig. 5 Stratification of rectal adenocarcinomas using the consensus molecular subtypes (CMS) classification.

(a) Expression levels for selected genes stratified by CMS group. Genes were annotated using the signatures from Budinska et al.70. (b) TMB stratified by CMS groups. Sample sizes are: CMS1 (n = 11), CMS2 (n = 26), CMS3 (n = 26), and CMS4 (n = 38). (c) FGA stratified by CMS groups. Sample sizes are: CMS1 (n = 11), CMS2 (n = 26), CMS3 (n = 26), and CMS4 (n = 38). (d) Percentage of KRAS mutated tumors by CMS group. (e) ssGSEA scores for selected pathways from the Hallmark dataset35. Sample sizes are: CMS1 (n = 11), CMS2 (n = 26), CMS3 (n = 26), and CMS4 (n = 38). (f) DFS for LARC patients treated with NAT, stratified by CMS group. (g) Levels of CA9 gene expression as a function of KRAS and PIK3CA mutational status. Double mutants and KRAS-mutant tumors had significantly higher expression of CA9 compared to wild-type tumors, p = 1.3e-07 and p = 4.65e-05, respectively. Sample sizes are: Double-mutant (n = 8), KRAS-mutant (n = 26), PIK3CA-mutant (n = 6), and wild-type (n = 5). Statistical significance was assessed using a two-sided Mann–Whitney U-test. (h) Expression of L1CAM stratified by CMS group. L1CAM expression was higher in CMS2 and CMS4 compared to CMS3, q = 0.0498 and q = 0.096, respectively. Sample sizes are: CMS1 (n = 11), CMS2 (n = 26), CMS3 (n = 26), and CMS4 (n = 38). (i) Validation of transcriptomic findings using an independent cohort of 15 LARC cases from Kamran et al.10 Differential gene expression was conducted using DESeq2 and the p-values attained by the Wald test were corrected using false discovery rates. In panels (B), (C), (E) and (H), statistical significance was assessed using a two-sided Mann–Whitney U-test. P values were corrected using the Bonferroni method and significant results are denoted as *q < 0.05, **q < 0.01, ***q < 0.005 and ****q < 0.001. In panels (B), (C), (E), (G), and (H), boxplots’ center lines indicate medians, edges indicate the interquartile range, and the whiskers extend to the highest and lowest values not considered outliers.

Extended Data Fig. 6 Supporting information for the characterization of immune hot pMMR/MSS LARC tumors with favorable outcomes from NAT.

(a) Quantification of intra-tumoral TILs from H&E slides for 20 patients, including cases from IG1 (n = 6), IG2 (n = 6), IG3 (n = 5) and IG4 (n = 3). Statistical significance was assessed using a two-sided Mann–Whitney U-test. P values were corrected using the Bonferroni method. Boxplots’ center lines indicate medians, edges indicate the interquartile range, and the whiskers extend to the highest and lowest values not considered outliers. Right panel shows correlation between estimated fractions of intra-tumoral and inter-tumoral TILs. Statistical significance was assessed using a two-sided Spearman correlation. Error bands represent 95% confidence intervals. (b) ssGSEA scores for immune cell signatures from Bindea et al.32. Displayed cell types are the ones with an adjusted p-value < 0.10 after Bonferroni correction, based on a Kruskal-Wallis test. (c) Comparison of ssGSEA scores for specific oncogenic pathway signatures from the Hallmark set35 across the four immune clusters. Displayed cell types are the ones with an adjusted p-value < 0.10 after Bonferroni correction, based on a Kruskal-Wallis test. In panels (B) and (C), sample sizes are: IG1 (n = 52), IG2 (n = 37), IG3 (n = 7), and IG4 (n = 5). (d) Correlation plot showing gene signatures for 27 selected oncogenic pathways (yellow diamonds) and immune cell infiltrates (green diamonds). Right panels show illustrative scatter plots for pairs of variables with strong positive and negative correlations. White dots in the correlation heatmap highlight pairs of variables with significant two-sided Spearman correlation after Bonferroni correction. Error bands represent 95% confidence intervals. In panels (B) and (C), statistical significance was assessed using a two-sided Mann–Whitney U-test. P values were corrected using the Bonferroni method and significant results are denoted as *q < 0.05, **q < 0.01, ***q < 0.005 and ****q < 0.001. Boxplots’ center lines indicate medians, edges indicate the interquartile range, and the whiskers extend to the highest and lowest values not considered outliers.

Extended Data Fig. 7 Validation of immune groups in an independent cohort of LARC tumors from TCGA.

Validation of results using an independent cohort of 42 LARC samples from TCGA. (a) Unsupervised hierarchical clustering of pMMR/MSS tumors using ssGSEA scores for a set of well established immune signatures reveals three groups with increasing levels of overall immune infiltrate (IG1–IG3). dMMR/MSI tumors were added later as a fourth group (IG4). (b) Tumors in IG4 had higher TMB and had lower FGA than tumors in the IG1–IG3 groups. Sample sizes for each group are as follows: IG1 (n = 16), IG2 (n = 17), IG3 (n = 7), and IG4 (n = 2). Boxplots’ center lines indicate medians, edges indicate the interquartile range, and the whiskers extend to the highest and lowest values not considered outliers. (c) Distribution of CMS classes across immune groups. (d) Selected significant differences in ssGSEA scores for specific immune cell types across immune groups. Sample sizes for each group are as follows: IG1 (n = 16), IG2 (n = 17), IG3 (n = 7), and IG4 (n = 2). (e) Comparison of expression levels for genes encoding proteins involved in immune checkpoint blockade. Sample sizes for each group are as follows: IG1 (n = 16), IG2 (n = 17), IG3 (n = 7), and IG4 (n = 2). In panels (D) and (E), statistical significance was assessed using a two-sided Mann–Whitney U-test. P values were corrected using the Bonferroni method and significant results are denoted as *q < 0.05, **q < 0.01, ***q < 0.005 and ****q < 0.001. Boxplots’ center lines indicate medians, edges indicate the interquartile range, and the whiskers extend to the highest and lowest values not considered outliers.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–11: Table S1, Clinical, histopathological, and sequencing data; Table S2: Summary of clinical characteristics for the full cohort. Table S3: Overview of cohorts and key clinicopathological features. Table S4: Summary of MutSigCV analysis using WES samples. Table S5: Sample identifiers and clinical information for cases in supplemental cohorts. Table S6: Summary of clinical characteristics for the treatment response cohort. Table S7: Summary of outcome analysis (response and DFS) using genomic data. Table S8: Summary of outcome analysis (response and DFS) using transcriptomic data. . Table S9: Results from TIL quantification analyses. Table S10: Validation of immune profiling results using data from TCGA. Table S11: List of genes on the MSK-IMPACT targeted sequencing panels.

Rights and permissions

Springer Nature or its licensor 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

Chatila, W.K., Kim, J.K., Walch, H. et al. Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer. Nat Med 28, 1646–1655 (2022). https://doi.org/10.1038/s41591-022-01930-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-01930-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer