This site is intended for
U.S.
Healthcare Professionals only.

For Patients
 

Discovering the Possibilities of I-O Biomarkers

With a focus on precision medicine, our research and development program aims to rapidly translate research into novel regimens to accelerate delivery of the right treatment, for the right patient, at the right time.

To accelerate our ability to identify precision medicine solutions for individual patients, BMS is pursuing a unique multifaceted approach to translational medicine. Based upon our comprehensive analysis of the tumor microenvironment, including tumor-intrinsic signaling and immune biology, BMS aims to identify clinical characteristics and Immuno-Oncology (I-O) biomarkers to determine the patient populations most likely to benefit from I-O therapy.

I-O biomarkers may be used to determine the immune potential of the tumor microenvironment

  • Research in the field of I-O biomarkers seeks to characterize the relationship between the immune system, the tumor and its microenvironment, and the host
  • Unique interactions among these factors contribute to the balance between activation and suppression of the antitumor immune response1-3
  • Tumors can be characterized based on their degree of immune-cell infiltration, ranging from noninflamed to inflamed4

I-O biomarkers that can identify inflamed tumors may help predict a pre-existing antitumor immune response.3,5

To identify I-O biomarkers that clarify this unique interplay between the immune system and the tumor, BMS biomarker research is focused on four key areas:

I-O biomarkers may be used to advance precision medicine, enabling tailored therapeutic solutions for individualized patients

  • For each patient, the interaction of the immune system, cancer, and therapy is complex and unique
  • Therefore, the goal of I-O biomarker development is to enable a more personalized approach to treatment by identifying patients who are likely to respond to specific immunotherapies3,5,8

BMS is committed to the exploration of biomarkers in I-O research. This includes the evaluation of multiple biomarkers, as a composite biomarker approach may provide a more accurate and comprehensive assessment of the tumor and tumor microenvironment.

Thumbnail for The role of I-O biomarkers and their role in immuno-therapy video Thumbnail for The role of I-O biomarkers and their role in immuno-therapy video

I-O Biomarkers: Under investigation for their role in immuno-therapy

See how several biomarkers are under investigation for their potential to predict response to immunotherapy

Watch video

Get I-O Resources

Order or download
educational tools for your
patients and practice

See all resources

Clinical Trials

Learn more about our
current clinical trials

Learn more

REFERENCES–Discovering the possibilities of I-O biomarkers

1. Gkretsi V, Stylianou A, Papageorgis P, Polydorou C, Stylianopoulos T. Remodeling components of the tumor microenvironment to enhance cancer therapy. Front Oncol. 2015;5:214. 2. Nelson D, Fisher S, Robinson B. The ‘‘Trojan Horse’’ approach to tumor immunotherapy: targeting the tumor microenvironment. J Immunol Res. 2014. doi:10.1155/2014/789069. 3. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348(6230):56-61. 4. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res. 2016;22(8):1865-1874. 5. Yuan J, Hegde PS, Clynes R, et al. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer. 2016;4:3. doi:10.1186/s40425-016-0107-3. 6. Blank CU, Haanen JB, Ribas A, Schumacher TN. The “cancer immunogram.” Science. 2016;352(6286):658-660. 7. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321-330. 8. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542-e551. 9. Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125(Pt 23):5591-5596. 10. Whiteside TL. Immune responses to cancer: are they potential biomarkers of prognosis? Front Oncol. 2013;3:1-8. 11. Ballman KV. Biomarker: predictive or prognostic? J Clin Oncol. 2015;33(33):3968-3971. 12. US Food and Drug Administration. About biomarkers. www.fda.gov/Drugs/DevelopmentApproval
Process/DrugDevelopmentToolsQualification
Program/BiomarkerQualificationProgram /ucm535922.htm. Accessed August 1, 2017. 13. Gainor JF, Longo DL, Chabner BA. Pharmacodynamic biomarkers: falling short of the mark? Clin Cancer Res. 2014;20(10):2587-2594. 14. Kluger HM, Zito CR, Barr ML, et al. Characterization of PD-L1 expression and associated T-cell infiltrates in metastatic melanoma samples from variable anatomic sites. Clin Cancer Res. 2015;21(13):3052-3060. 15. Hendry S, Salgado R, Gevaert T, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the IIBWG: Part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol. 2017;24(5):235-251. 16. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275-287. 17. Qiao M, Jiang T, Ren S, Zhou C. Combination strategies on the basis of immune checkpoint inhibitors in non-small-cell lung cancer: where do we stand? Clin Lung Cancer. 2018;19(1):1-11. 18. Cesano A, Warren S. Bringing the next generation of immuno-oncology biomarkers to the clinic. Biomedicines. 2018;6(1):1-11. 19. Kerr KM, Tsao MS, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol. 2015;10(7):985-989. 20. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124-128. 21. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69-74. 22. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015;21(1):24-33. 23. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016;9:5023-5039. 24. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847-856. 25. Van Allen EM, Wagle N, Levy MA. Clinical analysis and interpretation of cancer genome data. J Clin Oncol. 2013;31(15):1825-1833. 26. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949-954. 27. Mok TS. Personalized medicine in lung cancer: what we need to know. Nat Rev Clin Oncol. 2011;8(11):661-668. 28. Jakobsen JN, Santoni-Rugiu E, Ravn J, Sørensen JB. Intratumour variation of biomarker expression by immunohistochemistry in resectable non-small cell lung cancer. Eur J Cancer. 2013;49(11):2494-2503. 29. Henry NL, Hayes DF. Cancer biomarkers. Mol Oncol. 2012;6(2):140-146. 30. Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. 2010;5(6):463-466. 31. Yu SL, Xu LT, Qi Q, et al. Serum lactate dehydrogenase predicts prognosis and correlates with systemic inflammatory response in patients with advanced pancreatic cancer after gemcitabine-based chemotherapy. Sci Rep. 2017;7:45194. 32. Weide B, Elsässer M, Büttner P, et al. Serum markers lactate dehydrogenase and S100B predict independently disease outcome in melanoma patients with distant metastasis. Br J Cancer. 2012;107(3):422-428. 33. Allegra CJ, Jessup JM, Somerfield MR, et al. American Society of Clinical Oncology provisional clinical opinion: testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy. J Clin Oncol. 2009;27(12):2091-2096. 34. Sepulveda AR, Hamilton SR, Allegra CJ, et al. Molecular biomarkers for the evaluation of colorectal cancer: guideline from the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J Clin Oncol. 2017;35(13):1453-1486. 35. Cramer SD, Chang BL, Rao A, et al. Association between genetic polymorphisms in the prostate-specific antigen gene promoter and serum prostate-specific antigen levels. J Natl Cancer Inst. 2003;95(14):1044-1053. 36. Lilja H, Ulmert D, Vickers AJ. Prostate-specific antigen and prostate cancer: prediction, detection and monitoring. Nat Rev Cancer. 2008;8(4):268-278. 37. Gaudreau PO, Stagg J, Soulières D, Saad F. The present and future of biomarkers in prostate cancer: proteomics, genomics, and immunology advancements: supplementary issue: biomarkers and their essential role in the development of personalised therapies (A). Biomarkers in Cancer. 2016;8(S2):15-33. doi:10.4137/BIC.S31802. 38. Radich JP, Gooley T, Bryant E, et al. The significance of bcr-abl molecular detection in chronic myeloid leukemia patients “late,” 18 months or more after transplantation. Blood. 2001;98(6):1701-1707. 39. Yuda J, Miyamoto T, Odawara J, et al. Persistent detection of alternatively spliced BCR-ABL variant results in a failure to achieve deep molecular response. Cancer Sci. 2017;108(11):2204-2212. 40. Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer. 2005;5(3):172-183. 41. An X, Tiwari AK, Sun Y, Ding PR, Ashby CR, Chen ZS. BCR-ABL tyrosine kinase inhibitors in the treatment of Philadelphia chromosome positive chronic myeloid leukemia: a review. Leuk Res. 2010;34(10):1255-1268. 42. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227-258. 43. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.