Skip to main content

Advertisement

Log in

Tumor Heterogeneity Correlates with Less Immune Response and Worse Survival in Breast Cancer Patients

  • Breast Oncology
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

Intratumor heterogeneity implies that subpopulations of cancer cells that differ in genetic, phenotypic, or behavioral characteristics coexist in a single tumor (Ma in Breast Cancer Res Treat 162(1):39–48, 2017; Martelotto in Breast Cancer Res 16(3):210, 2014). Tumor heterogeneity drives progression, metastasis and treatment resistance, but its relationship with tumor infiltrating immune cells is a matter of debate, where some argue that tumors with high heterogeneity may generate neoantigens that attract immune cells, and others claim that immune cells provide selection pressure that shapes tumor heterogeneity (McGranahan et al. in Science 351(6280):1463–1469, 2016; McGranahan and Swanton in Cell 168(4):613–628, 2017). We sought to study the association between tumor heterogeneity and immune cells in a real-world cohort utilizing The Cancer Genome Atlas.

Methods

Mutant allele tumor heterogeneity (MATH) was calculated to estimate intratumoral heterogeneity, and immune cell compositions were estimated using CIBERSORT. Survival analyses were demonstrated using Kaplan–Meir curves.

Results

Tumors with high heterogeneity (high MATH) were associated with worse overall survival (p = 0.049), as well as estrogen receptor-positive (p = 0.011) and non-triple-negative tumors (p = 0.01). High MATH tumors were also associated with less infiltration of anti-tumor CD8 (p < 0.013) and CD4 T cells (p < 0.00024), more tumor-promoting regulatory T cells (p < 4e−04), lower expression of T-cell exhaustion markers, specifically PDL-1 (p = 0.0031), IDO2 (p = 0.34), ADORA2A (p = 0.018), VISTA (p = 0.00013), and CCR4 (p < 0.00001), lower expression of cytolytic enzymes granzyme A (p = 0.0056) and perforin 1 (p = 0.053), and low cytolytic activity score (p = 0.0028).

Conclusions

High heterogeneity tumors are associated with less immune cell infiltration, less activation of the immune response, and worse survival in breast cancer. Our results support the notion that tumor heterogeneity is shaped by selection pressure of tumor-infiltrating immune cells.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Ma D, Jiang YZ, Liu XY, Liu YR, Shao ZM. Clinical and molecular relevance of mutant-allele tumor heterogeneity in breast cancer. Breast Cancer Res Treat. 2017;162(1):39–48.

    Article  CAS  Google Scholar 

  2. Martelotto LG, Ng CK, Piscuoglio S, Weigelt B, Reis-Filho JS. Breast cancer intra-tumor heterogeneity. Breast Cancer Res. 2014;16(3):210.

    Article  Google Scholar 

  3. McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351(6280):1463–69.

    Article  CAS  Google Scholar 

  4. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613–28.

    Article  CAS  Google Scholar 

  5. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: A looking glass for cancer? Nat Rev Cancer. 2012;12(5):323–34.

    Article  CAS  Google Scholar 

  6. Andor N, Graham TA, Jansen M, et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med. 2016;22(1):105–13.

    Article  CAS  Google Scholar 

  7. Mroz EA, Rocco JW. MATH, a novel measure of intratumor genetic heterogeneity, is high in poor-outcome classes of head and neck squamous cell carcinoma. Oral Oncol. 2013;49(3):211–15.

    Article  CAS  Google Scholar 

  8. Rajput A, Bocklage T, Greenbaum A, Lee JH, Ness SA. Mutant-allele tumor heterogeneity scores correlate with risk of metastases in colon cancer. Clin Colorectal Cancer. 2017;16(3):e165–e170.

    Article  Google Scholar 

  9. PLOS Medicine Staff. Correction: intra-tumor genetic heterogeneity and mortality in head and neck cancer: Analysis of data from the cancer genome atlas. PLoS Med. 2015;12(6):e1001844.

    Article  Google Scholar 

  10. 10. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–45.

    Article  CAS  Google Scholar 

  11. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.

    Article  Google Scholar 

  12. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404.

    Article  Google Scholar 

  13. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1.

  14. Ramanathan R, Raza A, Sturgill J, et al. Paradoxical association of postoperative plasma sphingosine-1-phosphate with breast cancer aggressiveness and chemotherapy. Mediators Inflamm. 2017;2017:5984819.

    Article  Google Scholar 

  15. Ramanathan R, Olex AL, Dozmorov M, Bear HD, Fernandez LJ, Takabe K. Angiopoietin pathway gene expression associated with poor breast cancer survival. Breast Cancer Res Treat. 2017;162(1):191–98.

    Article  CAS  Google Scholar 

  16. Kim SY, Kawaguchi T, Yan L, Young J, Qi Q, Takabe K. Clinical relevance of microRNA expressions in breast cancer validated using the cancer genome atlas (TCGA). Ann Surg Oncol. 2017;24(10):2943–49.

    Article  Google Scholar 

  17. Narayanan S, Kawaguchi T, Yan L, Peng X, Qi Q, Takabe K. Cytolytic activity score to assess anticancer immunity in colorectal cancer. Ann Surg Oncol. 2018;25:2323–31

  18. Moro K, Kawaguchi T, Tsuchida J, et al. Ceramide species are elevated in human breast cancer and are associated with less aggressiveness. Oncotarget. 2018;9(28):19874–890.

    Article  Google Scholar 

  19. Kawaguchi T, Yan L, Qi Q, et al. Overexpression of suppressive microRNAs, miR-30a and miR-200c are associated with improved survival of breast cancer patients. Sci Rep. 2017;7(1):15945

    Article  Google Scholar 

  20. Young J, Kawaguchi T, Yan L, Qi Q, Liu S, Takabe K. Tamoxifen sensitivity-related microRNA-342 is a useful biomarker for breast cancer survival. Oncotarget. 2017;8(59):99978–989.

    Article  Google Scholar 

  21. Kawaguchi T, Yan L, Qi Q, et al. Novel MicroRNA-based risk score identified by integrated analyses to predict metastasis and poor prognosis in breast cancer. Ann Surg Oncol. 2018;25(13):4037–46.

    Article  Google Scholar 

  22. Kawaguchi T, Narayanan S, Takabe K. ASO author reflections: “From computer to bedside”: a new translational approach to immunogenomics. Ann Surg Oncol. 2018;25 Suppl 3:846–47.

    Article  Google Scholar 

  23. Rocco JW. Mutant allele tumor heterogeneity (MATH) and head and neck squamous cell carcinoma. Head Neck Pathol. 2015;9(1):1–5.

    Article  Google Scholar 

  24. Budczies J, Klauschen F, Sinn BV, et al. Cutoff finder: A comprehensive and straightforward web application enabling rapid biomarker cutoff optimization. PLoS One. 2012;7(12):e51862.

    Article  CAS  Google Scholar 

  25. Chang C, Hsieh MK, Chang WY, Chiang AJ, Chen J. Determining the optimal number and location of cutoff points with application to data of cervical cancer. PLoS ONE. 2017;12(4):e0176231.

    Article  Google Scholar 

  26. 26. Mazumdar M, Glassman JR. Categorizing a prognostic variable: Review of methods, code for easy implementation and applications to decision-making about cancer treatments. Stat Med. 2000;19(1):113-132.

    Article  CAS  Google Scholar 

  27. 27. Brondum L, Eriksen JG, Singers Sorensen B, et al. Plasma proteins as prognostic biomarkers in radiotherapy treated head and neck cancer patients. Clin Transl Radiat Oncol. 2017;2:46-52.

    Article  Google Scholar 

  28. Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–57

  29. Charoentong P, Finotello F, Angelova M, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18(1):248–62.

  30. Turashvili G, Brogi E. Tumor heterogeneity in breast cancer. Front Med (Lausanne). 2017;4:227.

    Article  Google Scholar 

  31. Morris LG, Riaz N, Desrichard A, et al. Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival. Oncotarget. 2016;7(9):10051–063.

    Article  Google Scholar 

  32. Ellsworth RE, Blackburn HL, Shriver CD, Soon-Shiong P, Ellsworth DL. Molecular heterogeneity in breast cancer: state of the science and implications for patient care. Semin Cell Dev Biol. 2017;64:65–72.

    Article  CAS  Google Scholar 

  33. Szulwach KE, Chen P, Wang X, et al. Single-cell genetic analysis using automated microfluidics to resolve somatic mosaicism. PLoS One. 2015;10(8):e0135007.

    Article  Google Scholar 

  34. Lindström LS, Yau C, Czene K, et al. Intratumor heterogeneity of the estrogen receptor and the long-term risk of fatal breast cancer. J Natl Cancer Inst. 2018;110(7):726–33.

    Article  Google Scholar 

  35. Roth A, Khattra J, Yap D, et al. PyClone: Statistical inference of clonal population structure in cancer. Nat Methods. 2014;11(4):396–98.

    Article  CAS  Google Scholar 

  36. Jiao W, Vembu S, Deshwar AG, Stein L, Morris Q. Inferring clonal evolution of tumors from single nucleotide somatic mutations. BMC Bioinformatics. 2014;15:35–2105-15-35.

  37. Oesper L, Mahmoody A, Raphael BJ. THetA: Inferring intra-tumor heterogeneity from high-throughput DNA sequencing data. Genome Biol. 2013;14(7):R80

    Article  Google Scholar 

  38. Carter SL, Cibulskis K, Helman E, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 2012;30(5):413–21.

    Article  CAS  Google Scholar 

  39. Gu-Trantien C, Loi S, Garaud S, et al. CD4(+) follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013;123(7):2873–92.

    Article  CAS  Google Scholar 

  40. Rajput AB, Turbin DA, Cheang MC, et al. Stromal mast cells in invasive breast cancer are a marker of favourable prognosis: A study of 4,444 cases. Breast Cancer Res Treat. 2008;107(2):249–57.

    Article  Google Scholar 

  41. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13(11):674–90.

    Article  CAS  Google Scholar 

  42. Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013;34(3):137–43.

    Article  CAS  Google Scholar 

  43. Cekic C, Linden J. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res. 2014;74(24):7239–49.

    Article  CAS  Google Scholar 

  44. Lines JL, Sempere LF, Broughton T, Wang L, Noelle R. VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res. 2014;2(6):510–17.

    Article  CAS  Google Scholar 

  45. Trapani JA, Smyth MJ. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2002;2(10):735–47.

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: KAM, TK, and KT. Development of methodology: TK, LY, and KT. Acquisition of data (acquired and managed patients, provided facilities, etc.): TK, QQ, XP, MA, and LY. Analysis and interpretation of data (e.g. statistical analysis, biostatistics, computational analysis): TK, QQ, XP, MA, and LY. Writing, review, and/or revision of the manuscript: KAM, TK, MO, JY, SP, EO, and KT. Administrative, technical, or material support (i.e. reporting or organizing data, constructing databases): QQ, XP, and LY. Study supervision: KT and TK.

Corresponding author

Correspondence to Kazuaki Takabe MD, PhD, FACS.

Ethics declarations

DISCLOSURES

Kazuaki Takabe is funded by the United States National Institute of Health – National Cancer Institute (R01CA160688) and Susan G. Komen Foundation (investigator-initiated research Grant (IIR12222224). This work was also supported by National Cancer Institute Grant P30CA016056 involving the use of Roswell Park Cancer Institute’s Bioinformatics and Biostatistics shared resources. Biospecimens or research pathology services for this study were provided by the Pathology Resource Network. Clinical Data Delivery and Honest Broker services for this study were provided by the Clinical Data Network. Kerry-Ann McDonald, Tsutomu Kawaguchi, Qianya Qi, Xuan Peng, Mariko Asaoka, Jessica Young, Mateusz Opyrchal, Li Yan, Santosh Patnaik, and Eigo Otsuji have no conflicts of interest to declare.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

FIG. S1

MATH level and patient survival for HER2-positive and -negative breast cancers in the (a) TCGA and (b) PAM50 cohorts. MATH mutant allele tumor heterogeneity, HER2 human epidermal growth factor receptor 2, TCGA The Cancer Genome Atlas (TIFF 288 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McDonald, KA., Kawaguchi, T., Qi, Q. et al. Tumor Heterogeneity Correlates with Less Immune Response and Worse Survival in Breast Cancer Patients. Ann Surg Oncol 26, 2191–2199 (2019). https://doi.org/10.1245/s10434-019-07338-3

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-019-07338-3

Navigation