Introduction
Over the past decade, immunotherapy has revolutionized the treatment of cancer. Immunotherapies targeted to immune checkpoint molecules such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have significantly improved the prognosis of patients with a variety of cancers.1 The discovery of CTLA-4’s function as an inhibitory molecule expressed on T cells in 19942,3 and the subsequent success of CTLA-4 checkpoint inhibition in clinical trials4–8 led to the Food and Drug Administration’s (FDA) approval of ipilimumab for the treatment of melanoma in 2011. More recently, inhibition of PD-1 and programmed death-ligand 1 (PD-L1) has been found to lead to durable tumor regression and prolonged disease stabilization in many types of solid tumors, including melanoma, non-small cell lung cancer (NSCLC), and renal-cell cancer.9–11 Two PD-1 inhibitors (nivolumab and pembrolizumab) and three PD-L1 inhibitors (atezolizumab, durvalumab, and avelumab) have been approved by the FDA for the treatment of various cancers.12 Finally, combined immune checkpoint blockade using nivolumab and ipilimumab has shown clinical efficacy in multiple cancer types.13,14
The use of immune checkpoint inhibitors must be closely monitored for severe immune-related adverse events,15 as treatment with immune checkpoint blockade is associated with 10% to 20% grade 3 or 4 toxicities.16 From a clinical perspective, then, the development of biomarkers to predict clinical response is critical to helping clinicians weigh the potential benefits of immunotherapy against its potential toxicities. Biomarkers may also accelerate the development of other immunotherapeutic therapies by identifying subpopulations in which these drugs would be most effective, thus allowing for clinical trial enrichment strategies.17
Biomarkers are biological indicators that can be subdivided into two categories: prognostic and predictive.18 While a prognostic biomarker indicates a patient’s disease outcome without treatment, a predictive biomarker indicates how a patient will respond to a given therapy and may itself be a target for therapy.19 Thus, a prognostic biomarker may help identify patients whose disease is high risk and who would benefit from aggressive therapy; a predictive biomarker may help identify patients who will benefit from a specific therapy.
In order to be effective and practical in the clinic setting, a biomarker must be both specific and sensitive, but must also be easy to use and cost-effective.20 Because it allows for assessment of the tumor immune microenvironment and is readily applied in the clinic, immunohistochemistry (IHC) has proven a powerful tool for the discovery and use of biomarkers.21–23 However, many IHC-based biomarkers have struggled to reach the clinic for a variety of reasons, in particular due to challenges with validation or inaccuracy in predicting outcome.24 A selection of key clinical and IHC-based biomarkers and the limitations of the use of IHC will be discussed further below.
Tumor-infiltrating lymphocytes
The tumor microenvironment (TME), composed of cell types including tumor-infiltrating lymphocytes (TILs),25 is increasingly implicated in a bidirectional interplay with tumor cells capable of promoting or preventing tumor growth and invasion.26 The recognition of such interactions has led to significant interest in TILs as a biomarker to both prognosticate disease outcomes and predict response to treatments such as immune checkpoint inhibitors. Early studies in primary melanoma identified the prognostic value of TILs using a classification of immune infiltrates as brisk, nonbrisk, or absent by conventional H&E staining.27 A higher density of TILs has been associated with favorable clinical outcomes in various cancer types, including breast cancer and melanoma.28,29 Immunohistochemistry has yielded further insight into the phenotypic characterization of these immune infiltrates. As melanocytic lesions progress from benign nevi to cutaneous malignant melanomas, the absolute number of TILs increases with a relative increase in the numbers of CD3+TIA-1+ resting cytotoxic T lymphocytes (CTLs) as compared to CD20+ B lymphocytes.30
In addition to its role as a prognostic biomarker, the density of TILs has also been described as a biomarker predictive of response to treatment. A 2014 study found that in HER2+ breast cancer patients treated with adjuvant trastuzumab, increased levels of TILs were correlated with decreased distant recurrence relative to patients receiving chemotherapy only.31 Another study in patients with breast cancer found that density of intratumoral lymphocytes as well as protein expression of CD3, CD20, and CXCR3 are significantly associated with pathologic complete response to neoadjuvant chemotherapy.32 Further, in melanoma, CD4+ and CD8+ lymphocyte infiltration predicts a statistically significant favorable response to anti-PD-1 immunotherapy, underscoring the role of IHC in predicting both disease outcomes and responses to therapy.33
CD8+ TILs and the Immunoscore
The density of CD8+ T cells in the tumor has been proposed as a more precise alternative to the density of TILs.34–36 In breast cancer, the use of IHC has demonstrated that total CD8+ lymphocytic density is an independent predictor of longer disease specific survival, and therefore that CD8+ T cell density can act as a prognostic biomarker.35,37 In metastatic melanoma, greater numbers of CTLs with CD8 staining have been shown to correlate with longer survival,38 a finding also demonstrated in colorectal and cervical cancer.39,40 Further, a study by Tumeh et al. found that high density of CD8+ TILs correlates with response to anti-PD-1 therapy in metastatic melanoma, thus suggesting that density of CD8+ TILs may also have a role as a predictive biomarker.41
As evidence increases that CD8+ TILs confer a favorable prognosis in a variety of solid tumors including ovarian, gallbladder, and NSCLC,34,42,43 the Immunoscore has been proposed as a method of classifying malignant tumors by quantifying the in situ immune cell infiltrates of two lymphocyte populations.34,42,43 In patients with colorectal cancer, the Immunoscore has been demonstrated as a biomarker with clinical utility in predicting disease recurrence following surgical resection and therefore in identifying patients likely to benefit from adjuvant therapy.44,45 The need to reinforce the prognostic and predictive value of the Immunoscore in other solid tumors as well as to identify follow-up parameters to modify its initial prognostic value will inevitably drive further biomarker research utilizing IHC.46
PD-1/PD-L1
PD-1 and PD-L1 are immune checkpoint molecules expressed on T cells, antigen-presenting cells, and tumor cells. Presence of PD1 or PDL1 have been proposed as biomarkers predictive of response to PD-1/PD-L1 blockade. These ligands inhibit T cell activity and are thus key to maintaining an immunosuppressed environment in the tumor. Both of these molecules are the target of a number of therapeutic antibodies intended to promote T cell activity within the tumor.47 Since the majority of tumors do not respond to PD-1/PD-L1 inhibition, PD-1/PD-L1 expression has been investigated as a potential biomarker for response. A study in patients with NSCLC treated with pembrolizumab found that patients for whom at least 50% of tumor cells expressed PD-L1 had a response rate of 45.2%, whereas for all the patients combined the response rate was 19.4%, thus suggesting that PD-L1 expression is a predictive biomarker for response to pembrolizumab and leading to FDA approval of pembrolizumab in NSCLC in the context of tumor-PD-L1 expression as a companion biomarker.48 Further, a meta-analysis found that PD-L1 expression on tumor and tumor-infiltrating immune cells is a predictor of response across tumor types.49 However, there remains disagreement in the field about whether PD-L1 expression alone is sufficient to accurately determine which patients will respond to checkpoint blockade. Indeed, a trial of stage III melanoma patients treated with pembrolizumab found that pembrolizumab was consistently effective both in patients with PD-L1-positive tumors and in patients with PD-L1-negative tumors, thus suggesting that PD-L1 is not a useful predictive biomarker in these patients.50
There are currently four IHC assays available to assess PD-L1 expression in patients who might be treated with anti-PD-L1 or anti-PD-1 in clinical trials. Three of these assays have shown consistency in direct comparisons , although the fourth assay indicates a lower PD-L1 expression in tumor and immune cells.51 There are several challenges with these IHC assays, namely intratumoral heterogeneity, variable temporal expression of PD-L1, and prohibitive pricing.52 As such, PD-L1 remains an unreliable predictive biomarker of response to PD-1/PD-L1 checkpoint inhibition.
Other predictive biomarkers
Although there is no definite biomarker predicting response to CTLA-4 checkpoint inhibition, several biomarkers have been proposed for this purpose. Higher protein levels of indoleamine 2,3 dioxygenase (IDO) and FoxP3 at baseline have been found to be associated with favorable clinical outcomes in patients treated with anti-CTLA-4 therapy.53 Other studies have highlighted the importance of the ratio of effector T cells to regulatory T cells within the tumor,54 with one study showing that the ratio of CD8+ effector T cells to FoxP3+ regulatory T cells is positively correlated with therapy-induced tumor necrosis in previously vaccinated cancer patients treated with anti-CTLA-4.55 Further, an increase from baseline of absolute lymphocyte counts was found to positively correlate with response to anti-CTLA-4 therapy.56 Broader changes of the immune response, such as an increase in T cell diversity, have also been noted to follow anti-CTLA-4 immunotherapy and to be associated with a higher response rate.57,58 Other biomarkers associated with response to anti-CTLA-4 therapy play a role only during or after treatment and, as such, cannot be used to predict response prior to therapy.59
Limitations of IHC for biomarker discovery and use
Despite IHC’s ubiquitous presence in research and diagnostic procedures, it suffers from several limitations; most notably, the lack of strict guidelines for staining often results in conflicting results among different institutions using different protocols and different antibodies.60,61 Indeed, McCabe et al. reported that different concentrations of HER2 antibody for staining could result in opposite prognostic implications for patients with breast cancer.60 Beyond antibody concentration consistency, numerous other components of IHC lack quality control. For example, whether an antibody binds to its target with adequate sensitivity and specificity is not routinely tested.62,63 The lack of staining reliability may also stem from the absence of quality control measures beyond the antibody itself. Variations in tissue fixation times, slide thickness, and antigen retrieval all impact the sensitivity and specificity of the antibodies used.64 As such, detailed and standardized protocols are necessary to allow systematic use IHC-based biomarkers.
Future directions
Automated IHC platforms have the potential to improve reliability and reproducibility of IHC, which so far has limited the use of IHC-based biomarkers in the clinic. Automated IHC platforms can be used in a clinical setting to create a “closed system” that prevents variations from being introduced.61 Further, automated image analysis platforms that decrease observer variability can more reliably quantitate biomarker positivity or negativity in patient samples.65 However, as of yet, these platforms remain only semi-automated as they require significant input from the user to aid in the machine learning process.
Although the biomarkers described in this review have been discovered and analyzed using traditional immunohistochemistry, new technologies allow for more sophisticated analyses of molecular markers. For example, technologies that allow for multiplexed immunofluorescence, such as Vectra® or AQUA, allow for analysis of multiple cell phenotypes at a time.66,67 Importantly, the multiplexing aspect of these technologies opens the possibility of evaluating the proximity between individual cells.68 This may allow for further specification of a biomarker. Indeed, our lab has recently used multiplexed immunofluorescence to find that a low CTL to macrophage ratio in the stroma is associated with lower overall survival, and that a closer distance of CTLs to HLA-DR− macrophages is associated with poor prognosis in melanoma.66 Such biomarker discovery has been facilitated by the use of multiplexed, quantitative IHC in many other tumor types, such as breast cancer, pancreatic cancer, and squamous cell cancer.68–70
Finally, while biomarkers may act as independent indicators, a single biomarker is often insufficient to clearly and safely stratify patients.71 Combining IHC with genomic and transcriptomic techniques may help in identification of more precise and predictive biomarkers, as many biomarkers have been discovered using these techniques.72,73 For example, Hugo et al. conducted genomic and transcriptomic analyses to define a subset of melanoma tumors with a specific transcriptomic signature (named IPRES) that are innately resistant to PD-1 checkpoint blockade.74 Ayers et al. discovered an IFN-γ-related gene expression profile that is consistent with T cell inflammation and that is an independent predictor of response to PD-1 blockade in nine cancers.75 Other studies have found that the tumor mutation burden is a strong predictor of response to immunotherapy in both melanoma and NSCLC.76,77 As such, while IHC-based techniques can be powerful on their own, combining these techniques with other assays, or further developing these techniques to become multiplexed and more quantitative, may help accelerate the discovery and validation of biomarkers.
KEY POINTS.
-
▪
Immunotherapy has successfully improved the prognosis of patients with cancer, but its use must be closely monitored for severe immune-related adverse events.
-
▪
Predictive and prognostic biomarkers allow clinicians to weigh the potential benefits of immunotherapy against its potential toxicities by stratifying patients into risk groups.
-
▪
Immunohistochemistry (IHC) is a powerful tool for the discovery and use of biomarkers, but current techniques are limited due to lack of reproducibility.
-
▪
The combination of IHC with genomic and transcriptomic analyses and the use of multiplexed and automated IHC both may help accelerate the discovery and validation of biomarkers.
SYNOPSIS.
Immunotherapy has drastically improved the prognosis of many patients with cancer, but it can also lead to severe immune-related adverse events. Biomarkers, which are molecular markers that indicate a patient’s disease outcome or a patient’s response to treatment, are therefore crucial to helping clinicians weigh the potential benefits of immunotherapy against its potential toxicities. Immunohistochemistry (IHC) has thus far been a powerful technique for discovery and use of biomarkers such as CD8+ tumor-infiltrating lymphocytes. However, IHC has limited reproducibility. Thus, if more IHC-based biomarkers are to reach the clinic, refinement of the technique using multiplexing or automation is key.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
DISCLOSURE STATEMENT
The authors disclose no potential conflicts of interest.
References
- 1.Weber J, Mandala M, Del Vecchio M, et al. : Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N Engl J Med 377:1824–1835, 2017 [DOI] [PubMed] [Google Scholar]
- 2.Walunas TL, Lenschow DJ, Bakker CY, et al. : CTLA-4 can function as a negative regulator of T cell activation. Immunity 1:405–13, 1994 [DOI] [PubMed] [Google Scholar]
- 3.Krummel MF, Allison JP: CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 182:459–65, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Weber J, Thompson JA, Hamid O, et al. : A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res 15:5591–8, 2009 [DOI] [PubMed] [Google Scholar]
- 5.Wolchok JD, Neyns B, Linette G, et al. : Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol 11:155–64, 2010 [DOI] [PubMed] [Google Scholar]
- 6.O’Day SJ, Maio M, Chiarion-Sileni V, et al. : Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: a multicenter single-arm phase II study. Ann Oncol 21:1712–7, 2010 [DOI] [PubMed] [Google Scholar]
- 7.Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. : Prolonged Survival in Stage III Melanoma with Ipilimumab Adjuvant Therapy. N Engl J Med 375:1845–1855, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hodi FS, O’Day SJ, McDermott DF, et al. : Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–23, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Topalian SL, Hodi FS, Brahmer JR, et al. : Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366:2443–54, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Patnaik A, Kang SP, Rasco D, et al. : Phase I Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients with Advanced Solid Tumors. Clin Cancer Res 21:4286–93, 2015 [DOI] [PubMed] [Google Scholar]
- 11.Brahmer JR, Tykodi SS, Chow LQ, et al. : Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366:2455–65, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gong J, Chehrazi-Raffle A, Reddi S, et al. : Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer 6, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wolchok JD, Kluger H, Callahan MK, et al. : Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369:122–33, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hellmann MD, Ciuleanu TE, Pluzanski A, et al. : Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med 378:2093–2104, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Harris SJ, Brown J, Lopez J, et al. : Immuno-oncology combinations: raising the tail of the survival curve. Cancer Biol Med 13:171–93, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Postow MA, Callahan MK, Wolchok JD: Immune Checkpoint Blockade in Cancer Therapy. J Clin Oncol 33:1974–82, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Freidlin B, Korn EL: Biomarker enrichment strategies: matching trial design to biomarker credentials. Nat Rev Clin Oncol 11:81–90, 2014 [DOI] [PubMed] [Google Scholar]
- 18.Nalejska E, Maczynska E, Lewandowska MA: Prognostic and predictive biomarkers: tools in personalized oncology. Mol Diagn Ther 18:273–84, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Oldenhuis CN, Oosting SF, Gietema JA, et al. : Prognostic versus predictive value of biomarkers in oncology. Eur J Cancer 44:946–53, 2008 [DOI] [PubMed] [Google Scholar]
- 20.Mabert K, Cojoc M, Peitzsch C, et al. : Cancer biomarker discovery: current status and future perspectives. Int J Radiat Biol 90:659–77, 2014 [DOI] [PubMed] [Google Scholar]
- 21.Wolff AC, Hammond ME, Hicks DG, et al. : Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 138:241–56, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Loupakis F, Pollina L, Stasi I, et al. : PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J Clin Oncol 27:2622–9, 2009 [DOI] [PubMed] [Google Scholar]
- 23.Howat WJ, Lewis A, Jones P, et al. : Antibody validation of immunohistochemistry for biomarker discovery: recommendations of a consortium of academic and pharmaceutical based histopathology researchers. Methods 70:34–8, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Diamandis EP: The failure of protein cancer biomarkers to reach the clinic: why, and what can be done to address the problem? BMC Med 10:87, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Oble DA, Loewe R, Yu P, et al. : Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun 9:3, 2009 [PMC free article] [PubMed] [Google Scholar]
- 26.Weber CE, Kuo PC: The tumor microenvironment. Surg Oncol 21:172–7, 2012 [DOI] [PubMed] [Google Scholar]
- 27.Clemente CG, Mihm MC Jr., Bufalino R, et al. : Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77:1303–10, 1996 [DOI] [PubMed] [Google Scholar]
- 28.Adams S, Gray RJ, Demaria S, et al. : Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol 32:2959–66, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mihm MC, Jr., Clemente CG, Cascinelli N: Tumor infiltrating lymphocytes in lymph node melanoma metastases: a histopathologic prognostic indicator and an expression of local immune response. Lab Invest 74:43–7, 1996 [PubMed] [Google Scholar]
- 30.Hussein MR, Elsers DA, Fadel SA, et al. : Immunohistological characterisation of tumour infiltrating lymphocytes in melanocytic skin lesions. J Clin Pathol 59:316–24, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Loi S, Michiels S, Salgado R, et al. : Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol 25:1544–50, 2014 [DOI] [PubMed] [Google Scholar]
- 32.Denkert C, Loibl S, Noske A, et al. : Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol 28:105–13, 2010 [DOI] [PubMed] [Google Scholar]
- 33.Uryvaev A, Passhak M, Hershkovits D, et al. : The role of tumor-infiltrating lymphocytes (TILs) as a predictive biomarker of response to anti-PD1 therapy in patients with metastatic non-small cell lung cancer or metastatic melanoma. Med Oncol 35:25, 2018 [DOI] [PubMed] [Google Scholar]
- 34.Sato E, Olson SH, Ahn J, et al. : Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 102:18538–43, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mahmoud SM, Paish EC, Powe DG, et al. : Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol 29:1949–55, 2011 [DOI] [PubMed] [Google Scholar]
- 36.Liu H, Zhang T, Ye J, et al. : Tumor-infiltrating lymphocytes predict response to chemotherapy in patients with advance non-small cell lung cancer. Cancer Immunol Immunother 61:1849–56, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liu S, Lachapelle J, Leung S, et al. : CD8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast Cancer Res 14:R48, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Erdag G, Schaefer JT, Smolkin ME, et al. : Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res 72:1070–80, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Galon J, Costes A, Sanchez-Cabo F, et al. : Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313:1960–4, 2006 [DOI] [PubMed] [Google Scholar]
- 40.Piersma SJ, Jordanova ES, van Poelgeest MI, et al. : High number of intraepithelial CD8+ tumor-infiltrating lymphocytes is associated with the absence of lymph node metastases in patients with large early-stage cervical cancer. Cancer Res 67:354–61, 2007 [DOI] [PubMed] [Google Scholar]
- 41.Tumeh PC, Harview CL, Yearley JH, et al. : PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515:568–71, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lin J, Long J, Wan X, et al. : Classification of gallbladder cancer by assessment of CD8(+) TIL and PD-L1 expression. BMC Cancer 18:766, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ameratunga M, Asadi K, Lin X, et al. : PD-L1 and Tumor Infiltrating Lymphocytes as Prognostic Markers in Resected NSCLC. PLoS One 11:e0153954, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Galon J, Mlecnik B, Bindea G, et al. : Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol 232:199–209, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pages F, Mlecnik B, Marliot 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 [DOI] [PubMed] [Google Scholar]
- 46.Galon J, Fox BA, Bifulco CB, et al. : Immunoscore and Immunoprofiling in cancer: an update from the melanoma and immunotherapy bridge 2015. Journal of Translational Medicine 14:273, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ott PA, Hodi FS, Robert C: CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res 19:5300–9, 2013 [DOI] [PubMed] [Google Scholar]
- 48.Garon EB, Rizvi NA, Hui R, et al. : Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372:2018–28, 2015 [DOI] [PubMed] [Google Scholar]
- 49.Khunger M, Hernandez AV, Pasupuleti V, et al. : Programmed Cell Death 1 (PD-1) Ligand (PD-L1) Expression in Solid Tumors As a Predictive Biomarker of Benefit From PD-1/PD-L1 Axis Inhibitors: A Systematic Review and Meta-Analysis. JCO Precision Oncology:1–15, 2017 [DOI] [PubMed] [Google Scholar]
- 50.Eggermont AMM, Blank CU, Mandala M, et al. : Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. N Engl J Med 378:1789–1801, 2018 [DOI] [PubMed] [Google Scholar]
- 51.Rimm DL, Han G, Taube JM, et al. : A Prospective, Multi-institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non-Small Cell Lung Cancer. JAMA Oncol 3:1051–1058, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mathew M, Safyan RA, Shu CA: PD-L1 as a biomarker in NSCLC: challenges and future directions. Ann Transl Med 5:375, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.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. Journal of Translational Medicine 9:204, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Quezada SA, Peggs KS, Curran MA, et al. : CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest 116:1935–45, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hodi FS, Butler M, Oble DA, et al. : Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A 105:3005–10, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Berman DM, Wolchok J, Weber J, et al. : Association of peripheral blood absolute lymphocyte count (ALC) and clinical activity in patients (pts) with advanced melanoma treated with ipilimumab. Journal of Clinical Oncology 27:3020–3020, 2009. 19470921 [Google Scholar]
- 57.Cha E, Klinger M, Hou Y, et al. : Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med 6:238ra70, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Postow MA, Manuel M, Wong P, et al. : Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J Immunother Cancer 3:23, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Manson G, Norwood J, Marabelle A, et al. : Biomarkers associated with checkpoint inhibitors. Ann Oncol 27:1199–206, 2016 [DOI] [PubMed] [Google Scholar]
- 60.McCabe A, Dolled-Filhart M, Camp RL, et al. : Automated quantitative analysis (AQUA) of in situ protein expression, antibody concentration, and prognosis. J Natl Cancer Inst 97:1808–15, 2005 [DOI] [PubMed] [Google Scholar]
- 61.O’Hurley G, Sjostedt E, Rahman A, et al. : Garbage in, garbage out: a critical evaluation of strategies used for validation of immunohistochemical biomarkers. Mol Oncol 8:783–98, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Saper CB: A guide to the perplexed on the specificity of antibodies. J Histochem Cytochem 57:1–5, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bordeaux J, Welsh A, Agarwal S, et al. : Antibody validation. Biotechniques 48:197–209, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Williams JH, Mepham BL, Wright DH: Tissue preparation for immunocytochemistry. J Clin Pathol 50:422–8, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Joshi AS, Sharangpani GM, Porter K, et al. : Semi-automated imaging system to quantitate Her-2/neu membrane receptor immunoreactivity in human breast cancer. Cytometry A 71:273–85, 2007 [DOI] [PubMed] [Google Scholar]
- 66.Gartrell RD, Marks DK, Hart TD, et al. : Quantitative Analysis of Immune Infiltrates in Primary Melanoma. Cancer Immunol Res 6:481–493, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Johnson DB, Bordeaux JM, Kim JY, et al. : Quantitative Spatial Profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 Predicts Improved Outcomes of anti-PD-1 Therapies in Metastatic Melanoma. Clin Cancer Res, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Carstens JL, Correa de Sampaio P, Yang D, et al. : Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun 8:15095, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ali HR, Dariush A, Provenzano E, et al. : Computational pathology of pre-treatment biopsies identifies lymphocyte density as a predictor of response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res 18:21, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Feng Z, Bethmann D, Kappler M, et al. : Multiparametric immune profiling in HPV-oral squamous cell cancer. JCI Insight 2, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Landers KA, Burger MJ, Tebay MA, et al. : Use of multiple biomarkers for a molecular diagnosis of prostate cancer. Int J Cancer 114:950–6, 2005 [DOI] [PubMed] [Google Scholar]
- 72.Wang K, Huang C, Nice EC: Proteomics, genomics and transcriptomics: their emerging roles in the discovery and validation of colorectal cancer biomarkers. Expert Rev Proteomics 11:179–205, 2014 [DOI] [PubMed] [Google Scholar]
- 73.Dijkstra KK, Voabil P, Schumacher TN, et al. : Genomics- and Transcriptomics-Based Patient Selection for Cancer Treatment With Immune Checkpoint Inhibitors: A Review. JAMA Oncol 2:1490–1495, 2016 [DOI] [PubMed] [Google Scholar]
- 74.Hugo W, Zaretsky JM, Sun L, et al. : Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165:35–44, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ayers M, Lunceford J, Nebozhyn M, et al. : IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127:2930–2940, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Van Allen EM, Miao D, Schilling B, et al. : Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350:207–211, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rizvi NA, Hellmann MD, Snyder A, et al. : Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348:124–8, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]