Abstract
Homing of tumor-specific cytotoxic T lymphocytes (CTLs) to the tumor tissues represents a vital step in procuring an effective anti-tumor immune response. Intratumoral accumulation of tumor-specific CTLs can be supported through local chemokine modulation using immune adjuvants or viral vectors, as well as vaccination, using peptide, protein or cell-based vaccines, including dendritic cell (DC) vaccines. Clinical and pre-clinical studies demonstrate that the current immunotherapy regimens are only effective when high numbers of CTLs are present within the tumor microenvironment (TME). Notably, many types of cancer take advantage of this principle and restrict T cell migration into the tumor, subverting the anti-tumor immune response and allowing uncontrolled tumor growth. This chapter discusses the mechanisms involved in the migration of CTLs into tumors and describes the feasible method of evaluating treatment-induced changes in the numbers of polyclonal tumor-specific CTLs in the TME and lymphoid tissues. The described method is widely applicable to multiple tumor models with wild-type antigen expression patterns, without the need for genetically-manipulated cancer cells or animals expressing defined T cell receptors.
Keywords: cancer, cytotoxic T lymphocyte (CTL), dendritic cell (DC), ELISpot, immunotherapy, murine models, oncology, tumor associated lymphocyte (TAL), tumor infiltrating lymphocyte (TIL), tumor microenvironment (TME)
Introduction
The accumulation of cytotoxic T cells (CTL) in the tumor microenvironments (TME) is an important prognostic factor for cancer patients and one of the key factors predicting the effectiveness of different forms of immunotherapy. Studies in multiple patient cohorts demonstrated that TME infiltration by CTL is uniformly predictive of prolonged survival (Fridman et al., 2011) in several cancer types including colorectal (Pages et al., 2005; Galon et al., 2007), ovarian (Zhang et al., 2003; Sato et al., 2005), bladder (Sharma et al., 2007), breast (Mahmoud et al., 2011; X. Li et al., 2019), melanoma (Azimi et al., 2012; Rahbar et al., 2015), pancreatic (Carstens et al., 2017). In contrast to the prognostically favorable CTLs (Pages et al., 2005; Galon et al., 2006; Anraku et al., 2008; Yamada et al., 2010), intratumoral regulatory T cells (Tregs) predict poor outcomes (Curiel et al., 2004; Clarke et al., 2006; Michel et al., 2008; Chaput et al., 2009), indicating therapeutic potential for selectively enhancing intratumoral CTL densities, relative to Tregs, or converting “cold” tumors into “hot” ones.
The accumulation of suppressive immune cells including Tregs (Shang et al., 2015), myeloid derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) are important to consider when evaluating the TME. These subsets of suppressive cells produce such suppressive factors as IL-10, TGFb, IDO, arginase and nitric oxide and can directly and indirectly inhibit the efficacy of ICI (Shang et al., 2015; Binnewies et al., 2018; Cassetta & Kitamura, 2018; Weber et al., 2018). In a clinical setting, frequency of circulating MDSCs is associated with a poor response to the ICI αCTLA-4 (Meyer et al., 2014; Weide et al., 2014; Martens et al., 2016; Sade-Feldman et al., 2016) and thus neutralization of this subset of cells has become an important topic of ongoing research (Weber et al., 2018). Further, suppressive cells can express the ligands for ICI including PD-L1/2 (PD-1) and CD80/86 (CTLA-4) (Kryczek et al., 2006; Kuang et al., 2009; Bloch et al., 2013).
The presence of tumor-specific T cells in the TME is particularly important for the ability of the individual patients to respond to immunotherapy with immune checkpoint inhibitors (ICI), such as the inhibitors of the programmed cell death-1 (PD-1) pathway, as demonstrated by the correlations between CTL accumulation and clinical outcomes of ICI (Ji et al., 2012; Herbst et al., 2014; Tumeh et al., 2014; Llosa et al., 2015). The presence of these cells, including tumor-infiltrating lymphocytes (TIL), immune cells that have exited the bloodstream and entered a solid tumor, and tumor associated lymphocytes (TAL), immune cells present within the intraperitoneal ascites of cancers including ovarian cancer, have the ability to combat the growing cancer once in proximity of the tumor (Matsuzaki et al., 2010; Huang et al., 2017). However, the systemic numbers of tumor-specific T cells in circulation are very poor predictors of clinical responses (Astsaturov et al., 2003; S. A. Rosenberg et al., 2005). This striking distinction highlights the key importance of CTL migration to the tumor site, in addition to their induction in the effectiveness of anticancer immunity. Thus, methods which promote CTL accumulation in or around the tumor have the potential to expand the benefit of ICI and potentially other cancer treatments to very large groups of the currently non-responsive patients.
While ICI is becoming the gold standard of care for many cancers, their effectiveness differs between different patient groups. While the response rates in melanoma (Brahmer et al., 2012; Hamid et al., 2013; Topalian et al., 2014; Luke & Ott, 2015), non-small cell lung cancer (Reck et al., 2016; Kanwal et al., 2018), bladder cancer (J. E. Rosenberg et al., 2016; Sharma et al., 2016; Bellmunt et al., 2017; Rouanne et al., 2018) and renal cell cancer (Topalian et al., 2012; Motzer et al., 2015; Weinstock & McDermott, 2015) can reach 40%, the overall response rates remain around the 15 – 20% (Xu-Monette et al., 2017; Ribas & Wolchok, 2018).
To enhance the percentage of the patients with clinical benefit, multiple combination therapies have been pursued (Atkins & Tannir, 2018; Johnson & Win, 2018; Tang et al., 2018), aiming to make “cold” tumors become “hot” (Trujillo et al., 2018). Three main types of interventions tested in this regard include activation of endogenous antigen presenting cells (APCs), direct intratumoral delivery of CTL-attracting chemokines and the modulation of the TME using different chemokine inducers and modulators.
Local activation of the stimulator of interferon genes (STING) pathway within the TME was shown to be an effective way of local DC activation and type-1 IFN production in mice, leading to T cell mediated anti-tumor immunity (Fuertes et al., 2013; Ohkuri et al., 2014; Woo et al., 2014). Alternative strategies include the use of vaccines which target exogenous antigens to DCs (Kreutz et al., 2013), ex vivo generated DC vaccines, such as Sipuleucel- T (Provenge®) (Kantoff et al., 2010; Fong et al., 2014), genetically-modified viruses, such as talimogene laherparepve (B. L. Liu et al., 2003; Ribas et al., 2017), or the delivery of chemokine genes such as CXCL11 (Francis et al., 2016; Z. Liu et al., 2016). Our alternative strategy, already being tested in early phase clinical trials include combinatorial chemokine modulating (CKM) regimen which can induce local intratumoral effect after either systemic or local administration (Obermajer et al., 2018) due to the enhanced susceptibility of the TME, compared to the surrounding healthy tissues (Muthuswamy et al., 2012). Examples of effective CKM combinations include type I IFNs combined with TLR ligands (Lu, 2014; Muthuswamy et al., 2015), IL-18 (Wong, Berk, et al., 2013; Wong, Muthuswamy, et al., 2013) and COX2 inhibitors (Wong et al., 2016; Theodoraki et al., 2018).
In order to facilitate preclinical evaluation of new therapeutic agents and their combinations effective in promoting intratumoral accumulation of tumor-specific CTLs, we developed a simple and generally applicable protocol to evaluate the numbers of tumor-specific CTLs in the TME, without the need for genetically-manipulated cells or animals, and cell tracers (Figure 1). In contrast to the existing alternatives, which use T cells from genetically manipulated mice with defined T cell receptors (such as OT-I), and genetically modified tumor cell lines which uniformly express highly immunogenic model antigens, such as ovalbumin (OVA) (reviewed in (Dranoff, 2011), our model evaluates the numbers of all T cells able to recognize tumor cells (rather than their selected antigens) and produce IFNγ in result of such recognition.
Figure 1: Key elements of the protocol.

Depending on the specific question, tumor-isolated TILs, or spleen-isolated cells, which may be further separated as CD4+ or CD8+ T cells, are rested overnights before testing of their specificity against the original cancer cells used in the tumor model of interest. The ELIspot plate(s) are processed after 24 hour incubation/co-culture. Placing sample from each mouse into 1 single column will easily organize your ELISpot plate for up to 12 experimental mice. TIL/TALs (Step 14) and splenocytes (Step 16) can be used across rows of the plate. Wells in which irradiated tumor cells are absent (Step 26) will serve as controls for each type of cell examined.
The ability to evaluate polyclonal T cell responses to multiple tumor-related weak antigens (mutated or over-expressed “self” antigens) eliminates the need for surrogate antigens or genetically-manipulated T cells with artificial TCRs, and allowing the evaluation of T cells recognizing different TAAs expressed by cancer cells. It eliminates the need for known cancer antigens and arbitrarily selected threshold of antigen specificity, reflected in the design of tetramers or defined TCRα/β combinations. Our protocol allows for enumeration of spontaneously-arising and immunotherapy-induced T cells which are able to recognize any cancer cell, without the need for prior identification of the relevant immunogenic epitopes. This eliminates any bias resulting from the selective evaluation of high affinity TCR-antigen interactions, and assures its immediate general applicability to multiple tumor models and mouse strains. We have tested this protocol with various murine tumor models including TIL extraction from MC38 tumors and TAL extraction from ID8 tumors (Obermajer et al., 2018), but anticipate that it can be used in multiple other tumor models.
Materials
Reagents
AN18 anti-IFNγ-coating antibody (Mabtech)
Phosphate buffered saline (PBS) (Gibco)
Ethanol 70% (vol/vol) diluted (EMD Serono)
RPMI-1640
- Cell culture medium (CCM) should be prepared ahead of TME. Supplement RPMI-1640 (Gibco) medium with:
- 1% (vol/vol) 100 U/ml penicillin-streptomycin (Gibco)
- 1% (vol/vol) 1 mM sodium pyruvate (Invitrogen)
- 1% (vol/vol) non-essential amino acids (NEAAs) (Gibco)
- 0.1% (vol/vol) 14.3 mM 2βME (Sigma-Aldrich)
- 1% (vol/vol) 2 mM L-glutamine (Gibco)
- 10% (vol/vol) FBS
- Digestion buffer is freshly prepared with reconstituted (according to manufacturer instructions) Mouse Tumor Dissociation Kit (Miltenyi) enzymes in the following relative amounts (can be scaled in 2.5 ml increments according to experiment size):
- 100 μl enzyme D
- 50 μl enzyme R
- 12.5 μl enzyme A
- 2337.5 μl CCM
ACK Lysis buffer (Life Technologies)
Trypan blue (0.4% (wt/vol)) solution (Corning Cellgro)
- 10x acidic (pH: 4.6. 1.051 g/ml) PBS is prepared with:
- 13.5 g NaCl
- 0.125 g Na2HPO4·2H2O
- 2.1 g KH2PO4
- 200 ml dH2O.
Standard Isotonic Percoll Solution (SIP) is freshly prepared by mixing Percoll with acidic PBS (see point 9 above) at 10:1 ratio.
60% (vol/vol), 45% (vol/vol) and 34% (vol/vol) SIP solutions are prepared for separation steps by diluting SIP with CCM
Fetal bovine serum (FBS) (Gibco) is stored in bottles direct from manufacturer at −20°C. Bottles are thawed slowly at 4°C and then heat inactivated for 30 min at 56°C. Heat-inactivated aliquots are stored at −20°C
Ethylenediaminetetraacetic acid (EDTA) (Life Technologies)
Wash buffer is made from PBS with 0.05% (vol/vol) Tween 20 (Fisher Scientific)
Bovine serum albumin (BSA) (Sigma-Aldrich)
R4–6A2 anti-IFNγ biotinylated detecting antibody (Mabtech) diluted in PBS/0.5% (wt/vol) BSA
ABC Reagent (Vector Laboratories)
ELISpot AEC substrate kit (Vector Laboratories)
dH2O (Life Technologies)
Mice/splenocytes
Tumor cells matching the cells used to inoculate the experimental mice are used as target cells in the ELISPOT assay
Mice bearing specific tumors are to be treated according to IACUC approved protocols
Equipment/Supplies (with examples of our current suppliers)
10, 200, 1000 μl Pipette tips (Ependorf)
20 – 200 μl multichannel pipette (Ependorf)
P10, P200, and P1000 Pipetman (Ependorf)
2, 5, 10 and 25 ml sterile, individually wrapped Pipettes (Falcon)
Pipet-Aid (Corning)
ELISpot plates (Millipore)
Parafilm (Bemis)
Scissors (Fisher Scientific)
Forceps (Fisher Scientific)
Scalpel (Bard-Parker)
Cell culture incubator set to 37°C and 5% (vol/vol) CO2 (Fisher Scientific)
70 and 100 μm cell strainers (Falcon)
3 ml syringe with the removable piston (Covidien or analogous)
15 and 50 ml polypropylene conical tubes
Centrifuge
Microscope
Hemocytometer
Gamma irradiator
ELISpot reader
Methods
Preparation I
-
1
Monitor experimental mice and continue to Step 2 when they are nearing experimental endpoint based on pre-determined IACUC approved parameters
-
2
Add 100 μl of 15 μg/ml AN18 and αIFN-γ coating antibody in PBS to each well of an ELISpot plate
-
3
Wrap the plate in parafilm and incubate overnight at 4°C
-
4
At experimental endpoint, euthanize mice in accordance to institutional guidelines
Tumor infiltrating lymphocytes (TIL)/Tumor associated lymphocyte (TAL) Isolation
-
5To harvest solid (i.e. transplantable subcutaneous, autochthonous, spontaneous) tumors and isolate TALs from each individual mouse:
- Sterilize the abdomen area of the mouse with 70% ethanol
- Using scissors make a small incision into the peritoneal cavity
- Using forceps and/or scissors collect the tumor tissue directly into a 15 ml conical tubes filled with 2 ml PBS
- Add ~2 ml of PBS onto a petri dish (cover the bottom) and transfer the tumor tissue
- Using two scalpels, mince the tissue to pieces <0.5 mm
- Transfer the tumor pieces to a new 15 ml tube and add ~4 ml of digestion buffer (cover the tumor pieces)
- Incubate tumors at 37°C for 30 min while rocking
- Wet a 100 μm strainer placed on top of 50 ml tube, transfer the tumor pieces onto the strainer
- Mash the tumor pieces with the piston of a 3 ml syringe
- Continuously wash with PBS to a final volume of ~20 ml
-
6To harvest tumor ascites (i.e. transplantable intraperitoneal) and isolate TAL from each individual mouse:
-
gSterilize the abdomen area of the mouse with 70% ethanol
-
hFill a 10 ml syringe with 5 ml PBS and use a 18G needle to inject the PBS into the lower left quadrant of the abdominal
-
iAspirate as much peritoneal fluid as possible being careful not to penetrate organs within the cavity
-
jTransfer the peritoneal fluid into 50 ml conical tube
-
g
Preparation of TIL/TAL for accessing T cell activity
-
11Centrifuge the TIL (Step 5) or TAL (Step 6) at 400 g for 3 – 5 min at room temperature
- Discard the supernatant and resuspend the cells in 1 ml ACK Lysis buffer to remove red blood cells
- Add an additional 3 ml of ACK Lysis Buffer on top and invert tube to mix
- Incubate the cells at room temperature for 4 min
- Add 30 ml of PBS and centrifuge the cells at 400 g for 3 min at room temperature
- Resuspend in 20 ml PBS and transfer over 70 μm strainer
- Count the cells using hemacytometer
- Centrifuge the cells at 400 g for 3 – 5 min at room temperature
- Resuspend the cells in 3 ml SIP (see “Reagents”; points 9–10) and transfer to a 15 ml tube
-
12
Slowly add 3 ml of 45% (vol/vol) SIP on top of the 60% (vol/vol) SIP layer by expelling the 45% (vol/vol) SIP at a shallow angle down the side of the tube
-
13
Slowly add 3 ml of 34% (vol/vol) SIP on top of the 45% (vol/vol) SIP layer by expelling the 34% (vol/vol) SIP at a shallow angle down the side of the tube
-
14
Centrifuge the tubes at 2,400g for 30 min at room temperature without applying the brake
-
15
Collect the bottom interface (~2–3 ml) into a new 15 ml conical tube and fill the tube with PBS
-
16
Centrifuge at 600g for 10 min at room temperature
-
17
Resuspend the cell pellet in 500 μl CCM
-
18
Adjust volume of TIL/TAL to 1 × 106/ml with CCM
Splenocyte Isolation
-
19Harvest spleen and isolate splenocytes from each individual mouse
- Sterilize the upper abdomen/chest area of the mouse with 70% ethanol
- Using scissors make a small incision into the left flank of the mouse
- Using forceps and/or scissors collect the spleen
- Wet a 100 μm strainer placed on top of 50 ml tube, transfer the spleen directly onto the strainer
- Fill a 10 ml syringe with PBS and using a 22G needle inject into the spleen
- Repeat 3 – 5x to thoroughly flush the cells from the spleen
- Mash the spleen with the piston of a 3 ml syringe
- Continuously wash with PBS to a final volume of ~20 ml
- Centrifuge the cells at 400 g for 3 – 5 min at room temperature
- Discard the supernatant and resuspend the cells in 1 ml ACK Lysis buffer to remove red blood cells
- Discard the supernatant and resuspend the cells in 1 ml ACK Lysis buffer to remove red blood cells
- Add an additional 3 ml of ACK Lysis Buffer on top and invert tube to mix
- Incubate the cells at room temperature for 4 min
- Add 30 ml of PBS and centrifuge the cells at 400 g for 3 min at room temperature
- Resuspend in 20 ml PBS and transfer over 70 μm strainer
- Count the cells using hemacytometer
-
20
Resuspend splenocytes at 5 × 106/ml in CCM
Preparation II
-
21
Wash the ELISpot plate (step 3) 3 TMEs with 200 μl RPMI-1640 for 5 minutes
-
22
Block plate with 200 μl CCM for 30 minutes at 37°C
-
23
Remove all CCM
-
24
Add 100 μl of 1 × 106 TIL/TAL (step 12; total 1 × 105) to 4 wells of the ELISpot plate (see Figure 1 for recommended plate setup)
-
25
Add 100 μl of 5 × 105 splenocytes (step 14; total 5 × 104) to 4 wells of the ELISpot plate
Preparation of cultured cancer cells as targets for assessing tumor-specificity of T cells
-
26
Harvest the relevant cancer cells from cultures, using 1 mM EDTA in PBS and centrifuge at room temperature
-
27
Resuspend cancer cells at 8 × 105 cells/ml CCM in a 50 ml conical tube
-
28
Irradiate with 20 Gy
-
29
To wash cancer cells, centrifuge at 400 g for 5 min at room temperature, resuspend in 20 ml PBS X2
-
30
Resuspend the cells at 4 × 105 cells/ml in CCM
-
31
Add 4 × 104 cancer cells in 100 μl of to each of 3 wells with TIL/TAL and/or 3 wells with splenocytes (triplicate co-cultures with the cancer cell targets)
-
32
Add 100 μl CCM to the remaining wells (TIL/TAL and/or splenocytes) for controls (triplicate control cultures).
ELISpot Procedure
-
33
Incubate plates at 37°C, 5% (vol/vol) C02 for 24 – 48 hours
-
34
Add 200 – 300 μl of wash buffer to each well for 5 minutes × 5
-
35
Add 100 μl of 1 μg/ml of diluted R4–6A2 biotinylated secondary antibody
-
36
Incubate at room temperature for 2 hours
-
37
About 10 min before the incubation is finished, prepare ABC reagent according to the manufacturer’s instructions
-
38
Add 200 – 300 μl of wash buffer to each well for 5 minutes × 5
-
39
Add 100 μl of ABC reagent, and incubate the plate at room temperature for 1 hour
-
40
Add 200 – 300 μl of wash buffer to each well for 5 minutes × 3
-
41
Add 200 – 300 μl of PBS to each well for 5 minutes × 2
-
42
While washing the plate, prepare AEC substrate according to the manufacturer’s instructions
-
43
Add 100 μl of AEC reagent per well.
-
44
Once spots are clearly visible (~5 – 20 minutes depending on assay) but before background appears, rinse the plate thoroughly with dH2O
-
45
Remove the plastic lid from the plate and allow the plate to dry completely.
-
46
Read the plate using an ELISpot reader according to manufacturer instructions
Notes
Based on our experiences with this protocol we would like to draw your attention to some critical issues to pay attention to during experimentation.
We anticipate this procedure to accurately evaluate IFN-γ producing T cells in the tumor site and lymphoid organs in mice with any type of tumor. Our methods have been optimized for the isolation of immune cells from both solid tumors and tumor ascites. Although, we have successfully used this approach to evaluate multiple subcutaneous and intraperitoneal tumors including MC38 and ID8 (Obermajer et al., 2018), we cannot eliminate the possibility that some tumors may require additional methods to prepare single cell suspension of tumor cells and isolate T cells.
We recommend harvesting the tumors well in advance of the animals succumbing to high tumor burden, in order to minimize the impact of secondary effects of high tumor burden (tumor ulceration, infection, metabolic changes) which may complicate the interpretation of data.
We recommend isolating the T cells from the largest amount of tumor tissue as possible, to account for intratumoral heterogeneity of T cell infiltrates.
While dissociating the tumor, if tumor pieces are very small they can be directly transferred to a 2 ml Eppendorf tube with ~500 μl digestion buffer. The entire contents of the tube can be transferred to the strainer.
An overnight rest period for all ex vivo cells used in this procedure including TIL, TAL, and splenocyte (Steps 14 and 16) is recommended.
Unrelated cultured cancer cells can be used as additional non-specific targets.
Depending on your model system, it may be advantageous to further enrich your splenocyte samples (Step 16) for CD4+ or CD8+ T cells using cell sorting or magnetic separation.
We have optimized the immune cell harvesting portion of this protocol for isolating total TILs or TALs from tumor-bearing mice. However, our protocol can be adapted to isolate other cell subsets by adding additional separating steps following Step 5/6. For example, to isolate Tregs additional steps to separate FoxP3+ by magnetic separation or flow cytometry based cell sorting methods could be included.
Although the production of IFN-γ is is known to be well correlated with CTL activity, our protocol allows inclusion of additional antibodies in Step 2, to evaluate alternative markers, such as TNF-α or Granzyme B.
Discussion: applications of the Protocol
This protocol is designed to enhance the numbers of tumor-specific CTLs within tumor lesions (and lymphoid tissues), and to evaluate their changing numbers in the course of immunotherapy. A wide range of examples is provided in our recent publication (Obermajer et al., 2018) and include DC vaccines and different forms of systemically- or locally applied modulation of TME, such as systemic or local application of combinatorial adjuvants, intratumoral injection of DCs or different viral therapies.
We anticipate that the presented protocol may be used to identify the effective combinations of immune activators with chemo- or radiotherapy, blockade of tumor survival pathways (Vanneman & Dranoff, 2012), oncolytic virus therapies (J. Li et al., 2011), TNF/TNF-R superfamily ligands (Jensen et al., 2010), and/or blockers of inhibitory factors in tumor microenvironment (e.g. ICI, IL-10, M-CSF, VEGF, IDO- or COX2 blockers), to prioritize the most promising approaches for extended mouse- and eventual clinical testing.
Acknowledgements
The preparation of the manuscript has been supported, by the Roswell Park-University of Pittsburgh Ovarian Cancer SPORE (1P50 CA159981) Roswell Park Seed Funds in support of the P01CA234212-A1, the Developmental Funds Award from the Roswell Park Alliance Foundation and the Rustum Family Foundation.
References
- Anraku M, Cunningham KS, Yun Z, Tsao MS, Zhang L, Keshavjee S, et al. (2008). Impact of tumor-infiltrating T cells on survival in patients with malignant pleural mesothelioma. J Thorac Cardiovasc Surg, 135(4), 823–829. doi: 10.1016/j.jtcvs.2007.10.026 [DOI] [PubMed] [Google Scholar]
- Astsaturov I, Petrella T, Bagriacik EU, de Benedette M, Uger R, Lumber G, et al. (2003). Amplification of virus-induced antimelanoma T-cell reactivity by high-dose interferon-alpha2b: implications for cancer vaccines. Clin Cancer Res, 9(12), 4347–4355. [PubMed] [Google Scholar]
- Atkins MB, & Tannir NM (2018). Current and emerging therapies for first-line treatment of metastatic clear cell renal cell carcinoma. Cancer Treat Rev, 70, 127–137. doi: 10.1016/j.ctrv.2018.07.009 [DOI] [PubMed] [Google Scholar]
- Azimi F, Scolyer RA, Rumcheva P, Moncrieff M, Murali R, McCarthy SW, et al. (2012). Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol, 30(21), 2678–2683. doi: 10.1200/JCO.2011.37.8539 [DOI] [PubMed] [Google Scholar]
- Bellmunt J, Powles T, & Vogelzang NJ (2017). A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: The future is now. Cancer Treat Rev, 54, 58–67. doi: 10.1016/j.ctrv.2017.01.007 [DOI] [PubMed] [Google Scholar]
- Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. (2018). Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med, 24(5), 541–550. doi: 10.1038/s41591-018-0014-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bloch O, Crane CA, Kaur R, Safaee M, Rutkowski MJ, & Parsa AT (2013). Gliomas promote immunosuppression through induction of B7–H1 expression in tumor-associated macrophages. Clin Cancer Res, 19(12), 3165–3175. doi: 10.1158/1078-0432.CCR-12-3314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. (2012). Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med, 366(26), 2455–2465. doi: 10.1056/NEJMoa1200694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carstens JL, Correa de Sampaio P, Yang D, Barua S, Wang H, Rao A, et al. (2017). Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun, 8, 15095. doi: 10.1038/ncomms15095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cassetta L, & Kitamura T (2018). Targeting Tumor-Associated Macrophages as a Potential Strategy to Enhance the Response to Immune Checkpoint Inhibitors. Front Cell Dev Biol, 6, 38. doi: 10.3389/fcell.2018.00038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaput N, Louafi S, Bardier A, Charlotte F, Vaillant JC, Menegaux F, et al. (2009). Identification of CD8+CD25+Foxp3+ suppressive T cells in colorectal cancer tissue. Gut, 58(4), 520–529. doi: 10.1136/gut.2008.158824 [DOI] [PubMed] [Google Scholar]
- Clarke SL, Betts GJ, Plant A, Wright KL, El-Shanawany TM, Harrop R, et al. (2006). CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS One, 1, e129. doi: 10.1371/journal.pone.0000129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. (2004). Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 10(9), 942–949. doi: 10.1038/nm1093 [DOI] [PubMed] [Google Scholar]
- Dranoff G (2011). Experimental mouse tumour models: what can be learnt about human cancer immunology? Nat Rev Immunol, 12(1), 61–66. doi: 10.1038/nri3129 [DOI] [PubMed] [Google Scholar]
- Fong L, Carroll P, Weinberg V, Chan S, Lewis J, Corman J, et al. (2014). Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer. J Natl Cancer Inst, 106(11). doi: 10.1093/jnci/dju268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francis L, Guo ZS, Liu Z, Ravindranathan R, Urban JA, Sathaiah M, et al. (2016). Modulation of chemokines in the tumor microenvironment enhances oncolytic virotherapy for colorectal cancer. Oncotarget. doi: 10.18632/oncotarget.7907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, & Kroemer G (2011). Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res, 71(17), 5601–5605. doi: 10.1158/0008-5472.CAN-11-1316 [DOI] [PubMed] [Google Scholar]
- Fuertes MB, Woo SR, Burnett B, Fu YX, & Gajewski TF (2013). Type I interferon response and innate immune sensing of cancer. Trends in Immunology, 34(2), 67–73. doi: 10.1016/j.it.2012.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, et al. (2006). Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science, 313(5795), 1960–1964. doi: 10.1126/science.1129139 [DOI] [PubMed] [Google Scholar]
- Galon J, Fridman WH, & Pages F (2007). The adaptive immunologic microenvironment in colorectal cancer: a novel perspective. Cancer Res, 67(5), 1883–1886. [DOI] [PubMed] [Google Scholar]
- Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. (2013). Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med, 369(2), 134–144. doi: 10.1056/NEJMoa1305133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. (2014). Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature, 515(7528), 563–567. doi: 10.1038/nature14011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang RY, Francois A, McGray AR, Miliotto A, & Odunsi K (2017). Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology, 6(1), e1249561. doi: 10.1080/2162402X.2016.1249561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen SM, Maston LD, Gough MJ, Ruby CE, Redmond WL, Crittenden M, et al. (2010). Signaling through OX40 enhances antitumor immunity. Seminars in Oncology, 37(5), 524–532. doi: 10.1053/j.seminoncol.2010.09.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, et al. (2012). An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother, 61(7), 1019–1031. doi: 10.1007/s00262-011-1172-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson CB, & Win SY (2018). Combination therapy with PD-1/PD-L1 blockade: An overview of ongoing clinical trials. Oncoimmunology, 7(4), e1408744. doi: 10.1080/2162402X.2017.1408744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. (2010). Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med, 363(5), 411–422. doi: 10.1056/NEJMoa1001294 [DOI] [PubMed] [Google Scholar]
- Kanwal B, Biswas S, Seminara RS, & Jeet C (2018). Immunotherapy in Advanced Non-small Cell Lung Cancer Patients: Ushering Chemotherapy Through the Checkpoint Inhibitors? Cureus, 10(9), e3254. doi: 10.7759/cureus.3254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kreutz M, Tacken PJ, & Figdor CG (2013). Targeting dendritic cells--why bother? Blood, 121(15), 2836–2844. doi: 10.1182/blood-2012-09-452078 [DOI] [PubMed] [Google Scholar]
- Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P, et al. (2006). B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med, 203(4), 871–881. doi: 10.1084/jem.20050930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, et al. (2009). Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med, 206(6), 1327–1337. doi: 10.1084/jem.20082173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, O’Malley M, Urban J, Sampath P, Guo ZS, Kalinski P, et al. (2011). Chemokine expression from oncolytic vaccinia virus enhances vaccine therapies of cancer. Mol Ther, 19(4), 650–657. doi: 10.1038/mt.2010.312 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Gruosso T, Zuo D, Omeroglu A, Meterissian S, Guiot MC, et al. (2019). Infiltration of CD8(+) T cells into tumor cell clusters in triple-negative breast cancer. Proc Natl Acad Sci U S A, 116(9), 3678–3687. doi: 10.1073/pnas.1817652116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, et al. (2003). ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther, 10(4), 292–303. doi: 10.1038/sj.gt.3301885 [DOI] [PubMed] [Google Scholar]
- Liu Z, Ravindranathan R, Li J, Kalinski P, Guo ZS, & Bartlett DL (2016). CXCL11-Armed oncolytic poxvirus elicits potent antitumor immunity and shows enhanced therapeutic efficacy. Oncoimmunology, 5(3), e1091554. doi: 10.1080/2162402X.2015.1091554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. (2015). The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov, 5(1), 43–51. doi: 10.1158/2159-8290.CD-14-0863 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu H (2014). TLR Agonists for Cancer Immunotherapy: Tipping the Balance between the Immune Stimulatory and Inhibitory Effects. Front Immunol, 5, 83. doi: 10.3389/fimmu.2014.00083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luke JJ, & Ott PA (2015). PD-1 pathway inhibitors: the next generation of immunotherapy for advanced melanoma. Oncotarget, 6(6), 3479–3492. doi: 10.18632/oncotarget.2980 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. (2011). Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol, 29(15), 1949–1955. doi: 10.1200/JCO.2010.30.5037 [DOI] [PubMed] [Google Scholar]
- Martens A, Wistuba-Hamprecht K, Geukes Foppen M, Yuan J, Postow MA, Wong P, et al. (2016). Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res, 22(12), 2908–2918. doi: 10.1158/1078-0432.CCR-15-2412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, et al. (2010). Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A, 107(17), 7875–7880. doi: 10.1073/pnas.1003345107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, et al. (2014). Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother, 63(3), 247–257. doi: 10.1007/s00262-013-1508-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michel S, Benner A, Tariverdian M, Wentzensen N, Hoefler P, Pommerencke T, et al. (2008). High density of FOXP3-positive T cells infiltrating colorectal cancers with microsatellite instability. Br J Cancer, 99(11), 1867–1873. doi: 10.1038/sj.bjc.6604756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. (2015). Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol, 33(13), 1430–1437. doi: 10.1200/JCO.2014.59.0703 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muthuswamy R, Berk E, Junecko BF, Zeh HJ, Zureikat AH, Normolle D, et al. (2012). NF-kappaB hyperactivation in tumor tissues allows tumor-selective reprogramming of the chemokine microenvironment to enhance the recruitment of cytolytic T effector cells. Cancer Res, 72(15), 3735–3743. doi: 10.1158/0008-5472.CAN-11-4136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muthuswamy R, Wang L, Pitteroff J, Gingrich JR, & Kalinski P (2015). Combination of IFNalpha and poly-I:C reprograms bladder cancer microenvironment for enhanced CTL attraction. J Immunother Cancer, 3, 6. doi: 10.1186/s40425-015-0050-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obermajer N, Urban J, Wieckowski E, Muthuswamy R, Ravindranathan R, Bartlett DL, et al. (2018). Promoting the accumulation of tumor-specific T cells in tumor tissues by dendritic cell vaccines and chemokine-modulating agents. Nat Protoc, 13(2), 335–357. doi: 10.1038/nprot.2017.130 [DOI] [PubMed] [Google Scholar]
- Ohkuri T, Ghosh A, Kosaka A, Zhu JZ, Ikeura M, David M, et al. (2014). STING Contributes to Antiglioma Immunity via Triggering Type I IFN Signals in the Tumor Microenvironment. Cancer Immunology Research, 2(12), 1199–1208. doi: 10.1158/2326-6066.Cir-14-0099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pages F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, et al. (2005). Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med, 353(25), 2654–2666. doi: 10.1056/NEJMoa051424 [DOI] [PubMed] [Google Scholar]
- Rahbar M, Naraghi ZS, Mardanpour M, & Mardanpour N (2015). Tumor-Infiltrating CD8+ Lymphocytes Effect on Clinical Outcome of Muco-Cutaneous Melanoma. Indian J Dermatol, 60(2), 212. doi: 10.4103/0019-5154.152571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. (2016). Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med, 375(19), 1823–1833. doi: 10.1056/NEJMoa1606774 [DOI] [PubMed] [Google Scholar]
- Ribas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, et al. (2017). Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell, 170(6), 1109–1119 e1110. doi: 10.1016/j.cell.2017.08.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ribas A, & Wolchok JD (2018). Cancer immunotherapy using checkpoint blockade. Science, 359(6382), 1350–1355. doi: 10.1126/science.aar4060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. (2016). Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet, 387(10031), 1909–1920. doi: 10.1016/S0140-6736(16)00561-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenberg SA, Sherry RM, Morton KE, Scharfman WJ, Yang JC, Topalian SL, et al. (2005). Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J Immunol, 175(9), 6169–6176. doi: 10.4049/jimmunol.175.9.6169 [DOI] [PubMed] [Google Scholar]
- Rouanne M, Roumiguie M, Houede N, Masson-Lecomte A, Colin P, Pignot G, et al. (2018). Development of immunotherapy in bladder cancer: present and future on targeting PD(L)1 and CTLA-4 pathways. World J Urol, 36(11), 1727–1740. doi: 10.1007/s00345-018-2332-5 [DOI] [PubMed] [Google Scholar]
- Sade-Feldman M, Kanterman J, Klieger Y, Ish-Shalom E, Olga M, Saragovi A, et al. (2016). Clinical Significance of Circulating CD33+CD11b+HLA-DR- Myeloid Cells in Patients with Stage IV Melanoma Treated with Ipilimumab. Clin Cancer Res, 22(23), 5661–5672. doi: 10.1158/1078-0432.CCR-15-3104 [DOI] [PubMed] [Google Scholar]
- Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. (2005). 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(51), 18538–18543. doi:0509182102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shang B, Liu Y, Jiang SJ, & Liu Y (2015). Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep, 5, 15179. doi: 10.1038/srep15179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma P, Callahan MK, Bono P, Kim J, Spiliopoulou P, Calvo E, et al. (2016). Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol, 17(11), 1590–1598. doi: 10.1016/S1470-2045(16)30496-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, et al. (2007). CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A, 104(10), 3967–3972. doi: 10.1073/pnas.0611618104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang J, Shalabi A, & Hubbard-Lucey VM (2018). Comprehensive analysis of the clinical immuno-oncology landscape. Ann Oncol, 29(1), 84–91. doi: 10.1093/annonc/mdx755 [DOI] [PubMed] [Google Scholar]
- Theodoraki MN, Yerneni S, Sarkar SN, Orr B, Muthuswamy R, Voyten J, et al. (2018). Helicase-Driven Activation of NFkappaB-COX2 Pathway Mediates theImmunosuppressive Component of dsRNA-Driven Inflammation in the Human Tumor Microenvironment. Cancer Res, 78(15), 4292–4302. doi: 10.1158/0008-5472.CAN-17-3985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. (2012). Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med, 366(26), 2443–2454. doi: 10.1056/NEJMoa1200690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. (2014). Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol, 32(10), 1020–1030. doi: 10.1200/JCO.2013.53.0105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trujillo JA, Sweis RF, Bao R, & Luke JJ (2018). T Cell-Inflamed versus Non-T Cell-Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection. Cancer Immunology Research, 6(9), 990–1000. doi: 10.1158/2326-6066.CIR-18-0277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. (2014). PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature, 515(7528), 568–571. doi: 10.1038/nature13954 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanneman M, & Dranoff G (2012). Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer, 12(4), 237–251. doi: 10.1038/nrc3237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, et al. (2018). Myeloid-Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune Checkpoint Inhibitors. Front Immunol, 9, 1310. doi: 10.3389/fimmu.2018.01310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weide B, Martens A, Zelba H, Stutz C, Derhovanessian E, Di Giacomo AM, et al. (2014). Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res, 20(6), 1601–1609. doi: 10.1158/1078-0432.CCR-13-2508 [DOI] [PubMed] [Google Scholar]
- Weinstock M, & McDermott D (2015). Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Ther Adv Urol, 7(6), 365–377. doi: 10.1177/1756287215597647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong JL, Berk E, Edwards RP, & Kalinski P (2013). IL-18-primed helper NK cells collaborate with dendritic cells to promote recruitment of effector CD8+ T cells to the tumor microenvironment. Cancer Res, 73(15), 4653–4662. doi: 10.1158/0008-5472.CAN-12-4366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong JL, Muthuswamy R, Bartlett DL, & Kalinski P (2013). IL-18-based combinatorial adjuvants promote the intranodal production of CCL19 by NK cells and dendritic cells of cancer patients. Oncoimmunology, 2(9), e26245. doi: 10.4161/onci.26245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong JL, Obermajer N, Odunsi K, Edwards RP, & Kalinski P (2016). Synergistic COX2 Induction by IFNgamma and TNFalpha Self-Limits Type-1 Immunity in the Human Tumor Microenvironment. Cancer Immunology Research, 4(4), 303–311. doi: 10.1158/2326-6066.CIR-15-0157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MYK, et al. (2014). STING-Dependent Cytosolic DNA Sensing Mediates Innate Immune Recognition of Immunogenic Tumors. Immunity, 41(5), 830–842. doi: 10.1016/j.immuni.2014.10.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu-Monette ZY, Zhang M, Li J, & Young KH (2017). PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front Immunol, 8, 1597. doi: 10.3389/fimmu.2017.01597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamada N, Oizumi S, Kikuchi E, Shinagawa N, Konishi-Sakakibara J, Ishimine A, et al. (2010). CD8+ tumor-infiltrating lymphocytes predict favorable prognosis in malignant pleural mesothelioma after resection. Cancer Immunol Immunother, 59(10), 1543–1549. doi: 10.1007/s00262-010-0881-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. (2003). Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med, 348(3), 203–213. doi: 10.1056/NEJMoa020177 [DOI] [PubMed] [Google Scholar]
