Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 26.
Published in final edited form as: Lancet. 2018 May 10;391(10135):2084–2086. doi: 10.1016/S0140-6736(18)30953-X

Immunoscore for (colorectal) cancer precision medicine

Prof Shuji Ogino 1,2,3,4, Marios Giannakis 4,5,6
PMCID: PMC5984170  NIHMSID: NIHMS961781  PMID: 29754776

Immunotherapies targeting immune checkpoints such as CTLA4, PDCD1 (programmed cell death 1, PD-1), and CD274 (PDCD1 ligand 1, PD-L1) have revolutionised oncology.1 High-level microsatellite instability (MSI-high) caused by mismatch repair deficiency is routinely used as a biomarker to predict response to immune checkpoint blockade.2 In colorectal carcinoma, MSI-high status correlates with tumour neoantigen loads, which in turn correlate with immune response.3 Combined analysis of tumoural and immune factors [e.g., so-called TIME (Tumour Immunity in the MicroEnvironment) classification] can be a new cancer classification system. However, immune response measurements have not been used as robust pathologic biomarkers in clinical practice. Along with the inherent complexity of the immune system, difficulty in reproducibly assessing immune cells remains a major cause of this gap.

In an ardent attempt to address the gap, the international immunoscore project 4 was initiated to standardise immune measurements, resulting in the study by Pagès et al.5 in this week's issue of Lancet. Pagès et al.5 assessed the reproducibility and prognostic role of the immunoscore assay that measured CD3+-cell and CD8+-cell densities in the tumour centre and invasive margin, using over 2,600 patients (51.5% male; a median age of 69 years, ranging 19-101) with stage I-III colon cancer across 13 countries. Standardised protocols for immunohistochemistry and digital image analysis generated highly reproducible results across institutions and operators (R > 0.96). The prognostication of the immunoscore was shown in both training and validation sets, and in time-to-recurrence (TTR, the primary endpoint), disease-free survival, and overall survival analyses. Using all cohorts, multivariable-adjusted hazard ratios (95% CI) in the TTR analysis were 0.65 (0.53-0.79) for intermediate (vs. low) immunoscores and 0.40 (0.30-0.54) for high (vs. low) immunoscores. Importantly, the immunoscore demonstrated a larger relative prognostic value than pT-stage, pN-stage, lymphovascular invasion, tumour differentiation, and MSI status. The prognostication by the tumour-node-metastasis (TNM) staging was improved with the addition of the immunoscore (TNM+I). Regarding a study limitation, approximately 10% of cases failed to satisfy specimen quality criteria, implying that immunoscore tests may not yield satisfactory results for some patients though recent evidence suggests a lower drop rate with the pre-defined standardised procedure.

The study by Pagès et al. 5 provides evidence for the immunoscore as a prognostic biomarker in colon cancer that can be standardised across pathology laboratories. The prognostic value of the immunoscore in colon cancer has been shown by an independent study.6 So what is next? There remain outstanding issues to resolve. Whether the immunoscore can serve as an actionable predictive biomarker for response to therapy remains to be investigated. Costs, benefits, and feasibility in pathology laboratory workflow need to be examined before implementing the immunoscore as a pathology test. In addition, T cells encompass very heterogenous cell populations, and other immune cells such as NK cells, dendritic cells, and macrophages are also important in determining behaviour of cancer cells. Technological advances including single cell transcriptome analysis (RNA-sequencing), multiplex immunofluorescence assays, and in vivo pathology technologies will allow better characterisation of tumour-immune interactions. There is still much to learn about the tumour-immune interactions and clinical utility of various existing and emerging immune biomarkers.

Factors other than tumour and immune cells must also be considered. Accumulating evidence points to the role of exposures (the exposome) in modulating tumour-immune interactions.7, 8 The microbiome is one such factor. Among all human organs, the colorectum has by far the most abundant amount of microorganisms. The gut and intratumor microbiota have been shown to affect responsiveness to chemotherapy and immunotherapy in gastrointestinal and other cancers.9 Recent molecular pathological epidemiology (MPE) research has provided evidence for influences of fibre-rich diets on the microbiota and carcinogenic process,10 and influences of omega-3 polyunsaturated fatty acids (PUFA) on tumour-immune interactions.11 Robust immune cell assays such as the immunoscore test can drive advancements of integrative immunology-MPE research, which can provide novel evidence for immunomodulatory roles of nutritional, lifestyle, and pharmacological factors including omega-3 PUFA, vitamin D, physical activity, and aspirin.8 Transdisciplinary science of immunology-MPE can also open opportunities to investigate the role of immune biomarkers in selecting patients for interventional use of an immunomodulatory factor.8 Although lagging behind immunotherapeutic advances, the development of cancer immunopreventive strategies is on the horizon.12, 13 Implementing immunomodulatory diets and lifestyle may be cost-effective cancer prevention strategies at the societal level.8

In summary, the study by Pagès et al.5 represents a great step toward standardised immune response measurements for solid tumours. After clinical utility and cost effectiveness are proven, the immunoscore assay can be implemented in clinical settings. The advancement of robust immune assays will upgrade population-scale cancer immunology research. Integrative analyses of exogenous and endogenous factors (including the microbiome) and tumour-immune interactions will enable even more personalised characterisation of cancer toward precision medicine. Widespread use of high-quality tumour-immune interaction analyses will transform clinical and population studies worldwide, which can eventually contribute to global cancer prevention and control.

Acknowledgments

Funding: This work was supported in part by grants from the USA National Institutes of Health (R35 CA197735 to SO), and Nodal Award (to SO) from the Dana-Farber Harvard Cancer Center.

Abbreviations

MPE

molecular pathological epidemiology

MSI

microsatellite instability

PD-1

programmed cell death 1

PUFA

polyunsaturated fatty acid

TIME

Tumor Immunity in the MicroEnvironment

TNM

tumour-node-metastasis

Footnotes

Competing interests: All authors declare that they have no conflicts of interest.

Use of standardised official symbols: We use Human Genome Organisation (HUGO)-approved official symbols (or root symbols) for genes and gene products, including CD3, CD8, CD274, CTLA4, and PDCD1; all of which are described at www.genenames.org. The official symbols are italicised, to differentiate from non-italicised colloquial names that are used along with the official symbols. This format enables readers to familiarise the official symbols for genes and gene products together with common colloquial names.

References

  • 1.Basile D, Garattini SK, Bonotto M, et al. Immunotherapy for colorectal cancer: where are we heading? Expert Opin Biol Ther. 2017;17(6):709–21. doi: 10.1080/14712598.2017.1315405. [DOI] [PubMed] [Google Scholar]
  • 2.Le DT, Hubbard-Lucey VM, Morse MA, et al. A Blueprint to Advance Colorectal Cancer Immunotherapies. Cancer Immunol Res. 2017;5(11):942–9. doi: 10.1158/2326-6066.CIR-17-0375. [DOI] [PubMed] [Google Scholar]
  • 3.Giannakis M, Mu XJ, Shukla SA, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 2016;15(no issue number):857–65. doi: 10.1016/j.celrep.2016.03.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the Immunoscore in the classification of malignant tumors. J Pathol. 2014;232(2):199–209. doi: 10.1002/path.4287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Pagès F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the prognostic classification of colon cancer. Lancet. 2018 doi: 10.1016/S0140-6736(18)30789-X. in press. [DOI] [PubMed] [Google Scholar]
  • 6.Wirta EV, Seppala T, Friman M, et al. Immunoscore in mismatch repair-proficient and -deficient colon cancer. J Pathol Clin Res. 2017;3(3):203–13. doi: 10.1002/cjp2.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zitvogel L, Pietrocola F, Kroemer G. Nutrition, inflammation and cancer. Nat Immunol. 2017;18(8):843–50. doi: 10.1038/ni.3754. [DOI] [PubMed] [Google Scholar]
  • 8.Ogino S, Nowak JA, Hamada T, et al. Integrative analysis of exogenous, endogenous, tumour, and immune factors for precision medicine. Gut. 2018 doi: 10.1136/gutjnl-2017-315537. in press published online on 6 February 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rajpoot M, Sharma AK, Sharma A, Gupta GK. Understanding the Microbiome: Emerging Biomarkers for Exploiting the Microbiota for Personalized Medicine against Cancer. Semin Cancer Biol. 2018 doi: 10.1016/j.semcancer.2018.02.003. in press published online on 6 February 2018. [DOI] [PubMed] [Google Scholar]
  • 10.Mehta RS, Nishihara R, Cao Y, et al. Association of Dietary Patterns With Risk of Colorectal Cancer Subtypes Classified by Fusobacterium Nucleatum in Tumor Tissue. JAMA Oncol. 2017;3(7):921–7. doi: 10.1001/jamaoncol.2016.6374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Song M, Nishihara R, Cao Y, et al. Marine omega-3 Polyunsaturated Fatty Acid Intake and Risk of Colorectal Cancer Characterized by Tumor-Infiltrating T Cells. JAMA Oncol. 2016;2(9):1197–206. doi: 10.1001/jamaoncol.2016.0605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fletcher R, Wang YJ, Schoen RE, Finn OJ, Yu J, Zhang L. Colorectal cancer prevention: Immune modulation taking the stage. Biochim Biophys Acta. 2018;1869:138–48. doi: 10.1016/j.bbcan.2017.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Spira A, Yurgelun MB, Alexandrov L, et al. Precancer Atlas to Drive Precision Prevention Trials. Cancer Res. 2017;77(7):1510–41. doi: 10.1158/0008-5472.CAN-16-2346. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES