T cell immunoreceptor with Ig and ITIM domains (TIGIT) |
|
-
•
Fourcade J, Sun Z, Chauvin JM, et al. CD226 opposes TIGIT to disrupt Tregs in melanoma[published online ahead of print, 2018 Jul 25]. JCI Insight. 2018;3(14):e121157.
|
|
CTLA-4 blockade |
Tumor infiltrating lymphocytes (TIL) |
|
-
•
Ruffini E, Asioli S, Filosso PL, et al. Clinical significance of tumor-infiltrating lymphocytes in lung neoplasms. Ann Thorac Surg 2009; 87: 365–371.
|
|
Absolute lymphocyte count (ALC) and absolute neutrophil count (ANC) |
|
-
•
Zaragoza J, Caille A, Beneton N, et al. High neutrophil to lymphocyte ratio measured before starting ipilimumab treatment is associated with reduced overall survival in patients with melanoma. Br. J. Dermatol., 174;2016:146–151.
|
|
|
Inducible co-stimulator (ICOS) |
|
-
•
Manson G, Norwood J, Marabelle A, et al. Biomarkers associated with checkpoint inhibitors. Annals of Oncology, Oxford University Press (OUP), 2016; 27 (7):1199–1206.
|
T cell repertoire (TCR) |
|
|
Tumor associated antigens (TAA) |
|
-
•
Weide B, Zelba H, Derhovanessian E, et al. Functional T cells targeting NY-ESO-1 or Melan-A are predictive for survival of patients with distant melanoma metastasis. J Clin Oncol. 2012;30(15):1835–41.
|
|
Myeloid derived suppressor cells (MDSC) |
|
-
•
Meyer C, Cagnon L, Costa-Nunes CM, et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother. 2014;63(3):247–57.
|
Regulatory T cells (Treg) |
|
-
•
deLeeuw RJ, Kost SE, Kakal JA, Nelson BH. The prognostic value of FoxP3+ tumor-infiltrating lymphocytes in cancer: a critical review of the literature. Clin Cancer Res. 2012;18(11):3022–9.
|
-
•
Hannani D, Vétizou M, Enot D, et al. Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Research. 2015;25(2):208–224.
|
|
|
Indoleamine 2,3-Dioxygenase (IDO) |
|
-
•
Hamid O, Schmidt H, Nissan A, et al. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med 2011;9:204.
|
Microbiome profile |
|
-
•
Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science (New York, NY). 2015;350(6264):1079–1084.
|
|
PD-1/PD-L1 blockade |
PD-L1 expression |
|
-
•
Patel, SP, Kurzrock, R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther April 12,015 (14) (4) 847–856.
|
|
-
•
Brody, R, Zhang, Y, Ballas, M, et al. PD-L1 expression in advanced NSCLC: Insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer. 2017 Oct;112:200–215.
|
ALC and ANC |
|
-
•
Lin G, Liu Y, Li S, et al. Elevated neutrophil-to-lymphocyte ratio is an independent poor prognostic factor in patients with intrahepatic cholangiocarcinoma. Oncotarget. 2016;7:50963–50971.
|
TIL |
|
-
•
Ruffini E, Asioli S, Filosso PL, et al. Clinical significance of tumor-infiltrating lymphocytes in lung neoplasms. Ann Thorac Surg 2009; 87: 365–371.
|
|
Peripheral blood markers |
|
-
•
Martens A, Wistuba-Hamprecht K, Foppen MG, et al. Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clinical cancer research: an official journal of the American Association for Cancer Research. 2016;22(12):2908–2918.
|
-
•
Krieg C, Nowicka M, Guglietta S, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;(2):144–153.
|
|
|
IDO |
|
-
•
McNamara MJ, Hilgart-Martiszus I, Barragan Echenique DM, et al. Interferon-gamma production by peripheral lymphocytes predicts survival of tumor-Bearing mice receiving dual PD-1/CTLA-4 blockade. Cancer Immunol. Res. 2016;21:650–657.
|
|
Mutational load |
-
•
High mutational load associated with positive clinical response in melanoma, NSCLC, bladder cancer, and colorectal cancer
|
-
•
Rooney MS, Shukla SA, Wu CJ, et al. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160: 48–61.
|
|
-
•
Hellmann MD, Callahan MK, Awad MM, et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell. 2018 May 14;33(5):853–861.e4.
|
Mismatch repair deficiency (mmrd) |
|
-
•
Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-Repair deficiency. N. Engl. J. Med. 2015;372:2509–2520.
|
-
•
Lee V, Murphy A, Le DT, Diaz Jr., LA. Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist. 2016;21:1200–1211.
|
TCR |
|
|
Microbiome profile |
|
-
•
Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97.
|
-
•
Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.
|
-
•
Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108.
|
-
•
Chowell D, Morris LGT, Grigg CM, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018; 2;359(6375):582–587.
|
|
-
•
High concentrations of Enterococcus faecium, Collinsella aerofaciens, Bifidobacterium adolescentis, Klebsiella pneumoniae, Veillonella parvula, Parabacteroides merdae, Lactobacillus sp., and Bifidobacterium longum commensal are associated with positive response to anti-PD-1 therapy
|
|
Human leukocyte antigen class I (HLA—I) genotype |
|
-
•
Snary, D. Barnstable, CJ, Bodmer, WF, et al. Molecular structure of human histocompatibility antigens: The HLA-C series. Eur. J. Immunol. 1977;7:580–585.
|
|
|
-
•
Marsh, SG, Parham, P, Barber, LD. The HLA Factsbook. Academic Press, 1999.
|
|
-
•
Bobisse S, Foukas PG, Coukos G, Harari A. Neoantigen-based cancer immunotherapy. Annals of Translational Medicine. 2016;4(14):262.
|
Mutational load and increased neoantigen (neoAg) frequency |
|
|
NeoAg-reactive CD4+ and CD8+ T cells |
|
-
•
Bobisse S, Foukas PG, Coukos G, Harari A. Neoantigen-based cancer immunotherapy. Annals of Translational Medicine. 2016;4(14):262.
|
NK cell frequency |
|
-
•
Böttcher JP, Bonavita E, Chakravarty P, et al. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell. 2018;172(5):1022–1037.e14.
|
Ki-67 Expression on PD-1+ CD8 T Cells |
|
-
•
Kamphorst AO, Pillai RN, Yang S, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114(19):4993–4998.
|
Signatures of T cell dysfunction and exclusion (TIDE) |
|
-
•
Jiang P, Gu S, Pan D, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018 Oct;24(10):1550–1558.
|
|
|
Anti-CD19 chimeric antigen receptor (CAR) T cell therapy |
Polyfunctional CAR T Cells |
|
-
•
Lin Y, Gallardo HF, Ku GY, et al. Optimization and validation of a robust human T-cell culture method for monitoring phenotypic and polyfunctional antigen-specific CD4 and CD8 T-cell responses. Cytotherapy. 2009;11(7):912–22.
|
IL-6/STAT3 |
|
-
•
Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018.
|
Upregulation of programs involved in effector differentiation, glycolysis, exhaustion, and apoptosis |
|
-
•
Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018.
|