Skip to main content
British Journal of Cancer logoLink to British Journal of Cancer
. 2017 Jul 13;117(4):451–460. doi: 10.1038/bjc.2017.220

HYPE or HOPE: the prognostic value of infiltrating immune cells in cancer

Tristan A Barnes 1, Eitan Amir 1,*
PMCID: PMC5558691  PMID: 28704840

Abstract

Interactions between immune and malignant cells have been known to have clinical relevance for decades. The potential for immune control is now being therapeutically enhanced with checkpoint inhibitors and other novel agents to improve outcomes in cancer. The importance of the immune infiltrate as a prognostic marker is increasingly relevant. In this minireview, we present an overview of the immune infiltrate and its spatial organisation, and summarise the prognostic value of immune cells in different cancer types. International collaborative efforts are standardising histopathologic reporting of the immune infiltrate, to allow application of these parameters in the clinical and research settings. In general terms, a ‘pro-inflammatory’ tumour microenvironment and infiltrating CD8-expressing T lymphocytes are associated with improved clinical outcomes in a broad range of tumour types. The inhibitory function of other immune cells, for example, myeloid-derived suppressor cells and regulatory T cells, appear to have a major role in disrupting the capacity for the immune control of cancers.

Keywords: immune cells, tumour microenvironment, prognostic markers, cancer


The immune system and malignant cells interact via a complex network. The importance of immune function in tumour development and control has been acknowledged for decades. As immunotherapy enters clinical practice, these underpinnings have more relevance as we try to identify predictive biomarkers for benefit from new therapies. The seminal papers describing the ‘hallmarks of cancer’ pronounced the capacity to avoid immune destruction as one of the requirements for malignancy (Hanahan and Weinberg, 2011). There is now a vast literature supporting immunosurveillance as a significant contributor to the natural history of malignancy. The interaction between tumours and the immune system has been described in three scenarios of ‘immunoediting’ (Schreiber et al, 2011); specifically: elimination (where immune surveillance successfully eradicates malignant cells); equilibrium (where the immune system exerts control over abnormal cells) and escape (where tumour cells evade immune mechanisms allowing growth and metastasis) (Mittal et al, 2014). In this mini-review, we aim to define the immune infiltrate and its spatial organisation as well as summarising the prognostic value of immune cells in different solid cancers.

Subtypes of immune cell infiltrate

Historically, studies have focused on the interaction between cytotoxic T lymphocytes and cancer cells. The role of other immune cells is also now recognised as contributing to the complex immune response in cancer, some of which promote tumour control and others facilitate cancer progression (Table 1; Figure 1).

Table 1. Cell types in the tumour immune infiltrate.

  Expression Function
Lymphoid
Cytotoxic T lymphocyte CD3, CD8 Recognise and lyse target cells through release of perforin and granzymes. Activated by dendritic cell antigen presentation via major histocompatibility complex (MHC) Class I antigen to T cell receptor. Apoptosis is induced in cells expressing specific antigen
Regulatory T lymphocyte (also called suppressor T cells) CD3, CD4, CD25, FOXP3 Serve to maintain tolerant to self antigens; inhibit dendritic cell function of antigen presentation and thus inhibits both the expansion and the differentiation of T effector cells. Polyclonal Tregs appear to modulate differentiation and cell trafficking
T helper lymphocyte (Th cell) CD4 Help to modulate immune responses. Activate and promote growth of cytotoxic T cells; maximise activity of phagocytes through interaction with MHC Class II; role in B cell antibody class switching (e.g., from immunoglobulin-M to immunoglobulin-G)
Natural killer cell (NK) CD16, CD56 A subset of cytotoxic lymphocytes that can be activated in the absence of MHC Class I antigen presentation, thus an important component of the innate immune system
Myeloid
Dendritic cells CD40 Act to process and present antigen on MHC Class I, and via co-stimulatory molecules they serve to activate T lymphocytes
Myeloid-derived suppressor cells (MDSC) CD11b, CD66b Pathologically activated immature myeloid cells, with morphological and phenotypical similarity to mononuclear and polymorphonuclear cells. Prevent activation of T cells and have a role in promoting tumour growth and metastasis
Macrophages CD68 Part of the innate immune defence with phagocytic capacity, and also have a role in adaptive immunity through activation of other immune cells via cytokine release. M1 macrophages are pro-inflammatory (largely driven by interferon (IFN)-gamma); M2 macrophages release anti-inflammatory cytokines such as IL4, IL10, TGF-beta and nuture tolerance

Figure 1.

Figure 1

Pathways affecting cytotoxic T lymphocyte activity within the tumour microenvironment.

Location and spatial organisation of the immune cell infiltrate

Although the distinction between peritumoural, stromal and intratumoural lymphocytes is made histopathologically (Table 2), this is likely an artificial segregation as this is a dynamic network that allows chemokine-generated cell movement between these areas.

Table 2. Location of the immune infiltrate.

Infiltrate Location
Intratumoural Within the mass (or nest) of malignant cells, with direct proximity between cancer and immune cells
Stromal In the surrounding connective tissues and blood vessels
Peritumoural Around the tumour and can refer to cells at the advancing margin of the tumour, in the stroma or the tissues adjacent to the tumour

A qualitative description of the interplay between tumour and immune cell infiltrate has been termed the ‘immune contexture’, and includes the location of specific immune cells, tertiary lymphoid structures (ectopic lymphoid aggregates that are generated during immune stimulation and exhibit structural characteristics of lymphoid organs), and the chemokines and cytokines involved in this microenvironmental organisation (Fridman et al, 2011). Methods for describing the immune infiltrate are a limitation of current pathological reporting. There remains a lack of consensus regarding reporting of tumour-infiltrating lymphocytes (TILs), including methods to subtype the infiltrating cells and their spatial organisation. International working groups are trying to create and validate reporting guidelines (Salgado et al, 2015). Immunophenotyping of the immune infiltrate by immunohistochemistry or immunofluorescence staining can be performed in tissue samples, or after generation of cell suspensions that are generated by mechanical or enzymatic breakdown of fresh tumour tissue (Stoll et al, 2015). The use of sectioned tissue specimens allows spatial understanding of cell position relative to tumour cells, however, similar to the use of cell suspension, is limited by challenges in antigen retrieval (capacity to bind identifying/specific proteins of interest) and poor standardisation. Novel approaches such as mRNA characterisation of immune co-regulated genes may help to identify and characterise the immune infiltrate (Stoll et al, 2015). Meta-analytical data suggest that in most cancers, the immune infiltrate is heterogeneous and there is limited reproducibility of leukocyte subtypes (Stoll et al, 2015). Although the presence of T cells is clearly important, the interplay between tumour antigens and major histocompatibility complex (MHC) molecules for antigen presentation is critical for efficient T cell activation. High affinity of the targeted peptides for MHC is required for strong stimulation of T cells to secrete cytokines and produce tumour eradication or control (Engels et al, 2013). The specific antigenicity of coding exons in mutated cancer genes is an area of research and the capacity to sequence whole genomes with greater speed and reduced cost is enhancing the capacity to identify potentially antigenic mutations.

Prognostic value of infiltrating immune cells

The prognostic value of lymphocytes in stromal, peritumoural and intratumoural locations remains unclear, with conflicting data from different tumour sites. Peritumoural lymphocytes at the advancing tumour margin and those in direct contact with tumour cells have been purported to carry the most prognostic weight particularly in some disease sites (see below). In general terms, a ‘pro-inflammatory’ tumour microenvironment and infiltrating CD8-expressing T lymphocytes are associated with improved clinical outcomes in a broad range of tumour types. In contrast, the inhibitory function of other immune cells, for example, myeloid-derived suppressor cells and regulatory T cells (Tregs) appear to have a major role in disrupting the capacity for the immune control of cancers and are therefore associated with worse outcome.

Perhaps counter-intuitively, favourable outcomes have also been observed in tumours infiltrated by inhibitory immune cells, for example, forkhead box P3-positive regulatory T cells (FOXP3) cells in colorectal cancer. This may represent a feedback loop in the context of an existing anti-tumour immune response and thus actually indicate increased tumour immunogenicity (Gajewski et al, 2013). Myeloid-derived suppressor cells (MDSC) and tumour-associated macrophages are both capable of negative regulation of innate and adaptive immune pathways. MDSCs have a role in tumour growth and metastasis via promotion of immune privilege (ability to tolerate the introduction of antigens without eliciting an inflammatory immune response), tumour microenvironment remodelling, establishment of a pre-metastatic niche (a scenario where non-cancer cells promote future metastasis) and interaction with tumour to promote differentiation, invasion and angiogenesis (Marvel and Gabrilovich, 2015). There is evidence that MDSC expansion is associated with more advanced stages of malignancy in multiple cancer types and also correlates with poor prognosis independent of tumour burden (Ugel et al, 2015). Paradoxically, anti-tumour immunity also leads to selective pressure on malignant cells, which ultimately leads to survival of tumour cells with reduced immunogenicity (Shankaran, 2001). There are also data supporting the hypothesis that tumour-infiltrating immune cells can promote invasion and metastases (Man et al, 2013), which may in part explain the heterogeneity of results between studies examining this topic.

Tumour-specific prognostic value

Breast cancer

In breast cancer, the presence of TILs is associated with improved prognosis in human epidermal growth factor receptor 2 (HER2) positive and triple negative breast cancers (TNBC), but not in luminal subtypes. In addition, the recognition of the prognostic value of the immune infiltrate has been the basis for establishing a breast cancer immunological grade (Salgado et al, 2015).

Independent of other clinicopathological prognostic factors or chemotherapy regimens, multiple studies have confirmed stromal TILs are associated with higher rates of pathological complete response (pCR) to neoadjuvant chemotherapy in all subgroups evaluated (including ER positive, HER2-positive tumours) (Dushyanthen et al, 2015). However, these differences in response only appear to translate into improved longer term outcomes in non-luminal tumours.

A meta-analysis of 25 published studies comprising over 22 000 patients, failed to show that immune infiltrates are associated with overall survival (OS) in unselected breast cancer patients, but did find such an association in TNBC (hazard ratio (HR): 0.79; 95% confidence interval (CI): 0.71–0.87). CD8-expressing lymphocytes were associated with improved disease-free survival (DFS; HR: 0.69; 95% CI: 0.56–0.84) and breast cancer-specific survival (HR: 0.78; 95% CI: 0.71–0.86) in the overall population, whereas the FOXP3-expressing lymphocytes were associated with worse DFS (HR: 1.47; 95% CI: 1.06–2.05) and OS (HR: 1.50; 95% CI: 1.15–1.97, P=0.004) (Mao et al, 2016).

Clinical trials have not reported an association between TIL, nuclear grade or histopathological grade in TNBC with most making the assumption that TNBC are high grade (Adams et al, 2014). It remains uncertain whether this association may be explained partly by response to chemotherapy; lower grade luminal tumours have lesser response to cytotoxic therapy and are less frequently associated with infiltrating immune cells.

A Th1 immune phenotype and mRNA profiles consistent with immune activation have also been associated with response to neoadjuvant chemotherapy (Denkert et al, 2015). There is more variability in results seen in trials reporting outcome for CD4-expressing T lymphocytes and FOXP3-expressing Tregs. The presence of Tregs prior to chemotherapy is associated with higher probability of attaining a pathological complete response (pCR), which probably reflects their association with a higher number of CD8-expressing cells. A high ratio of CD8:FOXP3 cells and a lower proportion of FOXP3 at the end of neoadjuvant chemotherapy may have a more meaningful prognostic value (Dushyanthen et al, 2015).

The current working group have recommended semi-quantitative assessment of stromal TILs and at this stage do not advocate for sub-classification of lymphocytes (Salgado et al, 2015). This is due to both the greater reproducibility of stromal TIL measurement compared with intratumoural TILs, which are difficult to distinguish from malignant cells in standard H&E sections, and the fact that in TNBC and HER2-positive breast cancer, the prognostic power of TILs persists among all subtypes of infiltrating immune cells (Salgado et al, 2015).

Colorectal cancer

Several scoring systems have been proposed for quantifying the inflammatory response in colorectal cancer. These include the Jass score, the Immunoscore and the Klintrup–Mäkinen grade of overall peritumoural inflammation (Park et al, 2014). There is evidence that TILs are associated with greater prognostic value than the American Joint Committee on Cancer TNM stage (Jochems and Schlom, 2011). In a meta-analysis of nine trials examining tumour inflammation in colorectal cancer, the pooled HR confirmed an OS benefit for patients with prominent TILs compared with those without, with a HR of 0.59 (95% CI: 0.48–0.72, P<0.001) and a HR for cancer-specific survival of 0.40 (95% CI: 0.27–0.61, P<0.001). There were differences between all the studies in the thresholds used to determine TIL positivity of tumours, for example, some used mean or median cut offs, others used high vs low scores of Klintrup–Mäkinen or Jass scores (Mei et al, 2014). The evaluation of T cell subsets and specific location of lymphocytic infiltrate did not show strong prognostic value, specifically CD3, CD8, FOXP3 and at different sites (tumour centre, peritumoural stroma and invasive tumour margin) were examined. CD3-positive cells at the invasive margin had OR for DFS of 0.4 (95% CI: 0.35–0.68) and for OS of 0.63 (95% CI: 0.42–0.93). This analysis was limited by significant inter-study heterogeneity (Mei et al, 2014). This contrasts to earlier individual study data showing statistically significant association between the type of immune cell density at the centre of the tumour or the infiltrating margin and patient outcome (Jochems and Schlom, 2011).

Ovarian cancer

In a meta-analysis of 10 studies comprising 1815 patients with treated ovarian carcinoma (Hwang et al, 2012), presence of intra-epithelial T lymphocytes was associated with improved OS (pooled HR for death 0.45, 95% CI: 0.34–0.58, P<0.001). CD3- and CD8-expressing lymphocytes were both examined, and both conferred a survival advantage; CD8 was examined more frequently and demonstrated a larger magnitude of effect on OS than CD3 (pooled HR: 0.46 and 0.57, respectively) (Hwang et al, 2012).

This positive association between CD8-expressing lymphocytes and clinical outcome is also observed in the assessment of patients before treatment and following neoadjuvant chemotherapy. Data on CD3-expressing lymphocytes, B cells and NK cells are less clear (Santoiemma and Powell, 2015). There are conflicting data regarding FOXP3-positive Tregs, with a few studies demonstrating superior outcome, but most studies suggesting a negative impact on survival outcomes through inhibition of cytotoxic T cell activity (Santoiemma and Powell, 2015). The measured absolute number of infiltrating cells may not be as important as the proportion of CD8-expressing cells relative to all infiltrating cells. The prognostic value of intratumoural CD8-positive lymphocytes appears superior even to the adequacy of surgical debulking in prognosticating for both progression free survival and OS (Zhang, 2003).

Non-small cell lung cancer

In a meta-analysis of 29 trials with over 86 000 patients, high levels of CD8-expressing cells infiltrating the tumour or in the tumour stroma of non-small cell lung cancer (NSCLC) specimens were associated with better OS (HR: 0.76 and 0.80, respectively) compared with tumours without lymphocytes present. CD3 expression also demonstrated similar findings; pooled HR for OS 0.65 (95% CI: 0.50–0.84, P=0.001) for stromal CD3 cells and 0.66 (95% CI: 0.45–0.97, P=0.03) for intratumoural CD3 cells. Presence of intratumoural CD4-expressing cells between the tumour cells resulted in improved OS (HR: 0.65; 95% CI: 0.46–0.91, P=0.01). Despite a higher effect size, a significant association between stromal CD4-expressing cells and outcome was not observed (HR 0.43; 95% CI: 0.07–2.61, P=0.36), likely due to greater heterogeneity. FOXP3-expressing T cells in the tumour stroma had association with worse progression-free and OS (HR: 2.14; 95% CI: 1.68–2.72; P<0.001) and 2.67 (95% CI: 1.74–4.08; P<0.001, respectively) (Geng et al, 2015).

Melanoma

Checkpoint inhibitors were first approved in melanoma after a long history of interest in the immune response to these tumours after observation of spontaneous responses (Mihm and Mule, 2015). One histopathological definition of the immune response in melanoma categorised the immune infiltrating response as ‘brisk’, a scenario where lymphocytes are demonstrated in the entire tumour mass or along the advancing edge; ‘non-brisk’, where lymphocytes are seen focally in the centre of the tumour or along part of the invasive margin; or ‘absent’ with no tumoural lymphocytes at all or lymphocytes seen, but not interacting with melanoma cells. These subgroups provide prognostic information in historical studies. In one study, melanoma-specific death was 30 and 50% lower in the non-brisk and brisk groups, respectively, compared with the absent group (Mihm and Mule, 2015). In contrast, studies report no survival advantage with lymphocytic infiltrate particularly with respect to tumours of earlier stage and not in the radial growth phase (Ladanyi, 2015). However, overall, there is a large body of evidence documenting the prognostic value of the immune infiltrate in melanoma (see summary in Table 3).

Table 3. Studies examining the prognostic impact of infiltrating immune cells in melanoma.

Study Number Result Association Cell type
(Kakavand et al, 2015) 60 Positive correlation between CD3, CD4 and CD8 cells in sentinel node and DFS/OS; PD1+ lymphocytes associated with worse outcome Positive association CD3, CD4 and CD8
(Saldanha et al, 2017) 655 Higher TILs are associated with better prognosis; confirms value of a simplified numerical TIL scoring system Positive association TIL
(Park and Kim, 2017) 177 Density of lymphocytes in the peritumoural and intratumoural regions were both prognostic Positive association TIL
(Obeid et al, 2016) 147 Expression of PD-L1 and PD-L2 correlated with increasing densities of immune cells. PD-L2 expression associated with improved OS Positive association PD-L2 and TILs
(Weiss et al, 2016) 1241 Melanomas with brisk TILs are defined by an immunostimulatory gene expression profile and improved prognosis compared with melanomas with non-brisk or absent TILs Positive association TILs
(Garg et al, 2016) 57 B cells are associated with a significantly better overall survival in patients with cutaneous primary melanomas of >1 mm Breslow depth Positive association B cells
(Bosisio et al, 2016) 710 Sheets/clusters of plasma cells associated with worse prognosis than melanomas without plasma cells Negative association Plasma cells
(Messaoudene et al, 2015) 39 NK cells in SLN associated with higher risk of relapse; NK cells did not correlate with thickness of primary but with patient age Negative association NK cells
(Fortes et al, 2015) 4133 High levels TILs associated with improved OS Positive association TIL
(Song et al, 2015) 82 TILs decreased the risk of distant metastases in oral mucosal melanoma Positive association TIL
(Donizy et al, 2015) 104 High levels TILs associated with improved OS Positive association TIL
(Eriksson et al, 2015) 4237 TILs demonstrated no prognostic value for survival No association TIL
(Thomas et al, 2013) 3330 High levels TILs associated with improved OS Positive association TIL
(Cintolo et al, 2013) 161 Absence of TIL was associated with worse DSS; In radial growth phase presence of TIL with regression was associated with a poor prognosis Positive association TIL
(Lee et al, 2013) 90 Brisk TILs were associated with improved prognosis in acral melanoma Positive association TIL
(Grotz et al, 2013) 250 TILs in elderly melanoma patients predicts both SLN metastasis and improved melanoma-specific outcomes Positive association TIL
(Azimi et al, 2012) 1865 TIL grade is an independent predictor of OS. Pronounced TIL infiltrate associated with excellent prognosis Positive association TIL
(Erdag et al, 2012) 147 Higher densities of CD8+ T cells correlated best with survival, a higher density of CD45+ leukocytes, T cells, and B cells also correlated with increased survival Positive association CD8, CD45, T cells and B cells
(Ladanyi et al, 2011) 106 CD20+ B cells most often found in peritumoural stroma, correlated with activated T lymphocytes and high number of these cells provided OS advantage Positive association CD20 B cells and activated T cells
(Knol et al, 2011) 102 High Foxp3 expression using qPCR predicts for worse progression free survival in stage III melanoma patients Negative association FOXP3
(Burton et al, 2011) 515 TIL response is a significant predictor of SLN metastasis but is not a major predictor of DFS or OS No association TIL

Abbreviations: DFS=disease-free survival; DSS=disease specific survival; NK=natural killer; OS=overall survival; SLN=sentinal lymph node; TIL=tumour-infiltrating lymphocytes.

Renal cell carcinoma

There is contradictory evidence regarding the role of the immune cell infiltrate in renal cell carcinoma. Multiple studies have demonstrated a worse outcome in patients with a neutrophilic, and/or lymphocytic infiltrate (Jochems and Schlom, 2011), a finding which appears reproducible (Table 4). The reasons for this are not clear.

Table 4. Immune cells in renal cell carcinoma.

Study Number patients Result Association Cell type
(Geissler et al, 2015) 104 Tumour-infiltrating NK cells and Th1 markers associated with increased OS, for example, HLA-DRC and CXCR3C T cells; whereas a high number of T cells, especially with high CD69 expression correlated with worse prognosis Positive association; negative association NK and Th1; T cells (CD69)
(Kang et al, 2013) 199 PD1-positive or FoxP3-positive lymphocytes predicted poor OS survival Negative association FOXP3, PD1+ lymphocytes
(Hotta et al, 2015) 105 Low levels of memory T cells had improved OS Negative association Memory T cells
(Eckl et al, 2012) 41 NK cell percentage does not provide prognostic information No association NK cells
(Liotta et al, 2011) 30 Increase in both peripheral and intratumoural Tregs associated with worse prognosis Negative association Tregs
(Li et al, 2009) 125 Increased peritumoural Tregs are associated with worse prognosis in clear cell renal cell carcinoma Negative association Tregs
(Bromwich et al, 2003) 73 Increased CD4+ T cells associated with worse cancer-specific survival; no association demonstrated with CD8+ T cells Negative association CD4

Abbreviations: NK=natural killer; OS=overall survival.

Head and neck cancer

Several clinical trials have demonstrated that tumour infiltration by CD3- and CD8-expressing T cells correlates with improved disease outcome in chemoradiotherapy-treated patients with head and neck cancer. This positive prognosis holds true regardless of the human papilloma virus (HPV) DNA status (Balermpas et al, 2016a). Smoking-associated tumours with higher degrees of genomic instability and higher antigenicity would be expected to have increased potential to activate an immune response; however, this is not supported by clinical evidence. There is conflicting information regarding differences in the immune infiltrate in HPV-positive vs negative status (Wansom et al, 2012; Partlova et al, 2015); see Table 5.

Table 5. Studies of TILs in head and neck squamous cell carcinoma.

Study Number patients Result Association Cell type
(Xu et al, 2017) 202 TIL level was an independent positive prognostic factor for DFS Positive association TIL
(Kogashiwa et al, 2017) 84 PD-L1 expression was associated with CD8+ tumour-infiltrating lymphocytes and better outcome in patients with locally advanced oropharyngeal SCC Positive association CD8
(Punt et al, 2016) 162 High number of T cells was correlated with improved DFS in HPV-positive oropharyngeal SCC; improved outcome correlated with active Th17 cells and lower IL-17(+) non-T cells Positive association T cells, Th17
(Nguyen et al, 2016) 278 Higher CD4 levels predicted improved OS and disease-specific survival Positive association CD4
(Caldeira et al, 2015) 28 Increased neutrophilic infiltration demonstrated in tumours with higher T stage; no correlation with survival No association Neutrophils
(Balermpas et al, 2016b) 161 CD8+ TILs constitute an independent prognostic marker in HNSCC patients treated with adjuvant chemoradiotherapy; prognostic benefit is apparent in HPV pos and neg subgroups Positive association CD8
(Partlova et al, 2015) 54 HPV-positive tumour showed significantly higher numbers of infiltrating IFNγ+ CD8+ T lymphocytes, IL-17+ CD8+ T lymphocytes, myeloid dendritic cells and are associated with better outcome compared to HPV-negative Positive association IFNγ+ CD8+ T cells, IL-17 CD8+ T cells
(Wolf et al, 2015) 39 CD68+ macrophages were found associated with positive nodes and poorer overall survival (not significant) Negative association Macrophages
(Ward et al, 2014) 270 TIL levels prognostic in HPV-positive HNSCC Positive association TIL
(Balermpas et al, 2014) 101 High infiltrating CD3+ and CD8+ cells correlate with survival outcomes with chemoradiation Positive association CD3+ and CD8+ T cells
(Nordfors et al, 2013) 203 Higher CD8(+) TIL counts correlated to a better 3-year OS in HPV pos; no correlation of CD4(+) TILs with survival outcomes Positive association; no association CD8; CD4
(Fraga et al, 2012) 70 CD57+ TILs do not correlate with survival outcomes No association CD57
(Wansom et al, 2012) 46 T-cell infiltration did not differ by HPV status; related to DSS and OS; after adjusting for HPV status, CD8, FoxP3, and total T cells were significantly associated with DSS and OS Positive association CD8, FOXP3 and total T cells
(Sun et al, 2012) 83 Tumour-infiltrating CD4+ CD25(high) Foxp3+ Tregs correlated with intratumoural COX-2 expression and were associated with a worse recurrence free survival in univariate but not multivariate analysis No association FOXP3
(Pretscher et al, 2009) 33 Intra-epithelial CD8 cells in metastatic lymph nodes and high CD20+ B cells in lymphoid tissue of lymph node metastases were associated with improved DFS Positive association CD8, B cells
(Badoual, 2006) 84 CD4+CD69+ T cells are associated with improved OS Positive association CD4CD69+ T cells

Abbreviations: DFS=disease-free survival; DSS = disease specific survival; HNSCC = head and neck SCC; OS=overall survival; SCC=squamous cell carcinoma; TIL=tumour-infiltrating lymphocytes.

Urothelial cancers

The approval of immunotherapy in the treatment of advanced urothelial malignancy suggests the relevance of the immune system. This is supported by most studies demonstrating the positive prognostic value of CD3, CD4 and CD8 T cells, and the negative association of FOXP3-positive T cells with survival, see Table 6.

Table 6. Studies examining prognostic impact of immune cells in bladder cancer.

Study Number patients Result Association Cell type
(Krpina et al, 2015) 115 CD3+ and CD8+ TIL are predictive of bladder cancer recurrence in patients with solitary low-grade non-muscle invasive bladder cancer Positive association CD3 and CD8
(Wang et al, 2015) 302 Intratumoural CD103(+) TILs inversely associated with tumour size. High CD103+ cells associated with improved OS. Positive association CD103+
(Zhang et al, 2015) 131 Tumour-infiltrating CD4(+) T cell density emerged as an independent prognostic factor for OS (HR: 2.75; P=0.004) Positive association CD4
Knief et al (2016) 149 FOXP3/CD8 (OS: P=0.013, HR: 1.32, 95% CIs: 1.06–1.65) ratios were significantly associated with briefer OS and time to cancer-specific death Negative association FOXP3/CD8 ratio
(Sjodahl et al, 2014) 296 CD3(+) TILs was significantly associated with good prognosis. Positive association with CD3 was modulated by CD68(+) TAMs. Strongest negative association with survival was a high ratio between CD68 and CD3 Positive association; Negative association CD3+ High CD68/CD3 ratio
(Sharma et al, 2007) 69 Higher numbers of CD8 TILs within the tumour (> or =8) had better DFS and OS Positive association CD8
(Hilmy et al, 2006) 103 No correlation between TIL level and prognosis No association TILs

Abbreviations: TAM=tumour-associated macrophage; OS=overall survival; TIL=tumour-infiltrating lymphocytes.

Hepatocellular carcinoma

Several studies have examined the role of the intratumoural and peritumoural (parenchymal) infiltrate in hepatocellular carcinoma (HCC) (Table 7). High levels of FOXP3 Tregs are associated with worse DFS and OS. Two large meta-analyses performed in 2014 demonstrate the importance of FOXP3 in both the development and prognosis of HCC (Huang et al, 2014; Zhao et al, 2014). Gabrielson et al, 2016 applied the Galon Immunoscore (Galon et al, 2014) to HCC and confirmed its prognostic value, CD3 and CD8 cell densities predicted recurrence with ORs of 5.8 (95% CI: 1.6–21.8) and 3.9 (95% CI: 1.1–14.2), respectively. PDL1 staining was positively correlated with high CD3 and CD8 density and predicted a lower rate of recurrence (Gabrielson et al, 2016). The applicability of these tools remains limited by routine access to technology to subtype these T cells.

Table 7. Studies examining prognostic value in HCC.

Study Number patients Result Association Cell type
(Sideras et al, 2017) 154 Low CD8+TIL associated with poor HCC-specific survival. Positive association CD8
(Gabrielson et al, 2016) 65 Intratumoural and peri-tumoural CD3+/CD8+ density associated with lower risk of recurrence Positive association CD3/CD8 ratio
(Tu et al, 2016) 57 FOXP3+ Tregs/CD4+ T cells ratio was an independent prognostic factor for OS Positive association FOXP3/CD4 ratio
(Wang et al, 2016) 66 Tumour CD4 and CD8 lower than non-neoplastic liver; high Foxp3 associated with poor OS, whereas low CD8 expression in non-neoplastic liver associated with high HCC recurrence rate. Negative association FOXP3
(He et al, 2015) 149 High neutrophil to lymphocyte ratio in peritumoural tissues correlated with poor prognosis in patients with HCC Negative association Neutrophil:lymphocyte ratio
(Sun et al, 2015) 449 CD8+ in tumour centre had highest prognostic impact on DFS and OS Positive association CD8
(Ozgur et al, 2014) 8 High FoxP3+ poorer DFS Negative association FOXP3
(Brunner et al, 2015) 119 IL-33 and CD8+ cells associated with prolonged OS Positive association IL-33 and CD8
(Huang et al, 2014) 1964 OS significantly lower in high FOXP3 infiltrated tumours than low (at 1, 3 and 5 years) (meta-analysis 13 studies) Positive association FOXP3

Abbreviations: HCC=hepatocellular carcinoma; OS=overall survival; TIL=tumour-infiltrating lymphocytes.

Other tumour types

The prognostic role of the immune infiltrate in less common malignancies is summarised in the Online Appendix.

Brain metastases

Although the central nervous system (CNS) has been purported to be an ‘immune privileged’ site, there is an increasing evidence supporting the role of immune infiltrating cells in brain tumours. In a study by Harter et al, TILs in brain metastases from different tumour types were quantified and associated with outcome. This was then validated in a breast cancer only brain metastases cohort. Carcinomas demonstrated more frequent stromal infiltration, whereas TILs in melanoma were more often diffusely infiltrative. High TILs level, high-programed cell death protein (PD)1+/CD8+ and programed death ligand (PDL)-1 staining were associated with smaller tumours but there was no significant association with survival demonstrated (Harter et al, 2015). In contrast, Bienkowski and Preusser, 2015 provide a review of the literature in which they concluded that tumour-infiltrating lymphocyte density in CNS metastases were strongly associated with improved OS.

Summary

Broadly speaking, the immune infiltrate can be classified as a ‘pro-inflammatory’ phenotype with infiltrating T cells and a cytokine profile consistent with immune activation. Immune control of tumours can occur spontaneously, and the presence of an immune infiltrate is generally a good prognostic sign. However, the immune infiltrate has variable effect in prognostic models depending on the tumour type, location of the cells and state of activation; the complexity of immune networks are likely oversimplified in current measurement models. Tumour evasion through inhibitory mechanisms may serve as a predictive marker for benefit from immunotherapy, which inhibits negative regulators of the immune system. Alternatively, the microenvironment may lack immune cell infiltration, and tumour resistance is likely through immune system ignorance (Gajewski et al, 2013), and therefore promoting immune activation is less likely to be successful in this setting.

For tumour-infiltrating immune cells to live up to the ‘hype’ of inducing and promoting long-term tumour control and contribute as valuable prognostic markers, their subtype (especially activated antigen specific cytotoxic T lymphocytes) and position (organised spatial response) need to be defined and measured in a standardised manner. Successful inclusion of immune cell markers in prognostic clinical models is becoming a realistic hope in some cancers.

Footnotes

The authors declare no conflict of interest.

References

  1. Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, Martino S, Wang M, Jones VE, Saphner TJ, Wolff AC, Wood WC, Davidson NE, Sledge GW, Sparano JA, Badve SS (2014) 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(27): 2959–2966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Azimi F, Scolyer RA, Rumcheva P, Moncrieff M, Murali R, McCarthy SW, Saw RP, Thompson JF (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] [PubMed] [Google Scholar]
  3. Badoual C (2006) Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res 12: 465–472. [DOI] [PubMed] [Google Scholar]
  4. Balermpas P, Michel Y, Wagenblast J, Seitz O, Weiss C, Rodel F, Rodel C, Fokas E (2014) Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br J Cancer 110(2): 501–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Balermpas P, Rödel F, Rödel C, Krause M, Linge A, Lohaus F, Baumann M, Tinhofer I, Budach V, Gkika E, Stuschke M, Avlar M, Grosu A-L, Abdollahi A, Debus J, Bayer C, Stangl S, Belka C, Pigorsch S, Multhoff G, Combs SE, Mönnich D, Zips D, Fokas E (2016. a) CD8+ tumour-infiltrating lymphocytes in relation to HPV status and clinical outcome in patients with head and neck cancer after postoperative chemoradiotherapy: a multicentre study of the German cancer consortium radiation oncology group (DKTK-ROG). Int J Cancer 138(1): 171–181. [DOI] [PubMed] [Google Scholar]
  6. Balermpas P, Rodel F, Rodel C, Krause M, Linge A, Lohaus F, Baumann M, Tinhofer I, Budach V, Gkika E, Stuschke M, Avlar M, Grosu AL, Abdollahi A, Debus J, Bayer C, Stangl S, Belka C, Pigorsch S, Multhoff G, Combs SE, Monnich D, Zips D, Fokas E (2016. b) CD8+ tumour-infiltrating lymphocytes in relation to HPV status and clinical outcome in patients with head and neck cancer after postoperative chemoradiotherapy: a multicentre study of the German cancer consortium radiation oncology group (DKTK-ROG). Int J Cancer 138(1): 171–181. [DOI] [PubMed] [Google Scholar]
  7. Bienkowski M, Preusser M (2015) Prognostic role of tumour-infiltrating inflammatory cells in brain tumours: literature review. Curr Opin Neurol 28(6): 647–658. [DOI] [PubMed] [Google Scholar]
  8. Bosisio FM, Wilmott JS, Volders N, Mercier M, Wouters J, Stas M, Blokx WAM, Massi D, Thompson JF, Scolyer RA, van Baren N, van den Oord JJ (2016) Plasma cells in primary melanoma. Prognostic significance and possible role of IgA. Mod Pathol 29(4): 347–358. [DOI] [PubMed] [Google Scholar]
  9. Bromwich EJ, McArdle PA, Canna K, McMillan DC, McNicol A, Brown M, Aitchison M (2003) The relationship between T-lymphocyte infiltration, stage, tumour grade and survival in patients undergoing curative surgery for renal cell cancer. Br J Cancer 89(10): 1906–1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brunner SM, Rubner C, Kesselring R, Martin M, Griesshammer E, Ruemmele P, Stempfl T, Teufel A, Schlitt HJ, Fichtner-Feigl S (2015) Tumor-infiltrating, interleukin-33-producing effector-memory CD8(+) T cells in resected hepatocellular carcinoma prolong patient survival. Hepatology 61(6): 1957–1967. [DOI] [PubMed] [Google Scholar]
  11. Burton AL, Roach BA, Mays MP, Chen AF, Ginter BA, Vierling AM, Scoggins CR, Martin RC, Stromberg AJ, Hagendoorn L, McMasters KM (2011) Prognostic significance of tumor infiltrating lymphocytes in melanoma. Am Surg 77(2): 188–192. [PubMed] [Google Scholar]
  12. Caldeira PC, de Andrade Sousa A, de Aguiar MC (2015) Differential infiltration of neutrophils in T1-T2 versus T3-T4 oral squamous cell carcinomas: a preliminary study. BMC Res Notes 8: 569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cintolo JA, Gimotty P, Blair A, Guerry D, Elder DE, Hammond R, Elenitsas R, Xu X, Fraker D, Schuchter LM, Czerniecki BJ, Karakousis G (2013) Local immune response predicts survival in patients with thick (t4) melanomas. Ann Surg Oncol 20(11): 3610–3617. [DOI] [PubMed] [Google Scholar]
  14. Denkert C, Gv Minckwitz, Brase JC, Sinn BV, Gade S, Kronenwett R, Pfitzner BM, Salat C, Loi S, Schmitt WD, Schem C, Fisch K, Darb-Esfahani S, Mehta K, Sotiriou C, Wienert S, Klare P, André F, Klauschen F, Blohmer J-U, Krappmann K, Schmidt M, Tesch H, Kümmel S, Sinn P, Jackisch C, Dietel M, Reimer T, Untch M, Loibl S (2015) Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2–positive and triple-negative primary breast cancers. J Clin Oncol 33(9): 983–991. [DOI] [PubMed] [Google Scholar]
  15. Donizy P, Kaczorowski M, Halon A, Leskiewicz M, Kozyra C, Matkowski R (2015) Paucity of tumor-infiltrating lymphocytes is an unfavorable prognosticator and predicts lymph node metastases in cutaneous melanoma patients. Anticancer Res 35(1): 351–358. [PubMed] [Google Scholar]
  16. Dushyanthen S, Beavis PA, Savas P, Teo ZL, Zhou C, Mansour M, Darcy PK, Loi S (2015) Relevance of tumor-infiltrating lymphocytes in breast cancer. BMC Med 13: 202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Eckl J, Buchner A, Prinz PU, Riesenberg R, Siegert SI, Kammerer R, Nelson PJ, Noessner E (2012) Transcript signature predicts tissue NK cell content and defines renal cell carcinoma subgroups independent of TNM staging. J Mol Med 90(1): 55–66. [DOI] [PubMed] [Google Scholar]
  18. Engels B, Engelhard Victor H, Sidney J, Sette A, Binder David C, Liu Rebecca B, Kranz David M, Meredith Stephen C, Rowley Donald A, Schreiber H (2013) Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell 23(4): 516–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Erdag G, Schaefer JT, Smolkin ME, Deacon DH, Shea SM, Dengel LT, Patterson JW, Slingluff CL (2012) Immunotype and immunohistologic characteristics of tumor infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res 72(5): 1070–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Eriksson H, Frohm-Nilsson M, Järås J, Kanter-Lewensohn L, Kjellman P, Månsson-Brahme E, Vassilaki I, Hansson J (2015) Prognostic factors in localized invasive primary cutaneous malignant melanoma: results of a large population-based study. Br J Dermatol 172(1): 175–186. [DOI] [PubMed] [Google Scholar]
  21. Fortes C, Mastroeni S, Mannooranparampil TJ, Passarelli F, Zappala A, Annessi G, Marino C, Caggiati A, Russo N, Michelozzi P (2015) Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 25(4): 306–311. [DOI] [PubMed] [Google Scholar]
  22. Fraga CA, de Oliveira MV, Domingos PL, Botelho AC, Guimaraes AL, Teixeira-Carvalho A, Correa-Oliveira R, De Paula AM (2012) Infiltrating CD57+ inflammatory cells in head and neck squamous cell carcinoma: clinicopathological analysis and prognostic significance. Appl Immunohistochem Mol Morphol 20(3): 285–290. [DOI] [PubMed] [Google Scholar]
  23. Fridman WH, Galon J, Pagès F, Tartour E, Sautès-Fridman C, Kroemer G (2011) Prognostic and Predictive Impact of Intra- and Peritumoral Immune Infiltrates. Cancer Res 71(17): 5601–5605. [DOI] [PubMed] [Google Scholar]
  24. Gabrielson A, Wu Y, Wang H, Jiang J, Kallakury B, Gatalica Z, Reddy S, Kleiner D, Fishbein T, Johnson L, Island E, Satoskar R, Banovac F, Jha R, Kachhela J, Feng P, Zhang T, Tesfaye A, Prins P, Loffredo C, Marshall J, Weiner L, Atkins M, He AR (2016) Intratumoral CD3 and CD8 T-cell densities associated with relapse-free survival in HCC. Cancer Immunol Res 4(5): 419–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Gajewski TF, Schreiber H, Fu Y-X (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14(10): 1014–1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C (2014) Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol 232: 199–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Garg K, Maurer M, Griss J, Bruggen MC, Wolf IH, Wagner C, Willi N, Mertz KD, Wagner SN (2016) Tumor-associated B cells in cutaneous primary melanoma and improved clinical outcome. Hum Pathol 54: 157–164. [DOI] [PubMed] [Google Scholar]
  28. Geissler K, Fornara P, Lautenschlager C, Holzhausen HJ, Seliger B, Riemann D (2015) Immune signature of tumor infiltrating immune cells in renal cancer. Oncoimmunology 4(1): e985082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Geng Y, Shao Y, He W, Hu W, Xu Y, Chen J, Wu C, Jiang J (2015) Prognostic role of tumor-infiltrating lymphocytes in lung cancer: a meta-analysis. Cell Physiol Biochem 37(4): 1560–1571. [DOI] [PubMed] [Google Scholar]
  30. Grotz TE, Vaince F, Hieken TJ (2013) Tumor-infiltrating lymphocyte response in cutaneous melanoma in the elderly predicts clinical outcomes. Melanoma Res 23(2): 132–137. [DOI] [PubMed] [Google Scholar]
  31. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5): 646–674. [DOI] [PubMed] [Google Scholar]
  32. Harter PN, Bernatz S, Scholz A, Zeiner PS, Zinke J, Kiyose M, Blasel S, Beschorner R, Senft C, Bender B, Ronellenfitsch MW, Wikman H, Glatzel M, Meinhardt M, Juratli TA, Steinbach JP, Plate KH, Wischhusen J, Weide B, Mittelbronn M (2015) Distribution and prognostic relevance of tumor-infiltrating lymphocytes (TILs) and PD-1/PD-L1 immune checkpoints in human brain metastases. Oncotarget 6(38): 40836–40849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. He G, Zhang H, Zhou J, Wang B, Chen Y, Kong Y, Xie X, Wang X, Fei R, Wei L, Chen H, Zeng H (2015) Peritumoural neutrophils negatively regulate adaptive immunity via the PD-L1/PD-1 signalling pathway in hepatocellular carcinoma. J Exp Clin Cancer Res 34: 141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hilmy M, Campbell R, Bartlett JMS, McNicol AM, Underwood MA, McMillan DC (2006) The relationship between the systemic inflammatory response, tumour proliferative activity, T-lymphocytic infiltration and COX-2 expression and survival in patients with transitional cell carcinoma of the urinary bladder. Br J Cancer 95(9): 1234–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hotta K, Kato Y, Leighl N, Takigawa N, Gaafar RM, Kayatani H, Hirata T, Ohashi K, Kubo T, Tabata M, Tanimoto M, Kiura K (2015) Magnitude of the benefit of progression-free survival as a potential surrogate marker in phase 3 trials assessing targeted agents in molecularly selected patients with advanced non-small cell lung cancer: systematic review. PLoS One 10(3): e0121211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Huang Y, Liao H, Zhang Y, Yuan R, Wang F, Gao Y, Wang P, Du Z (2014) Prognostic value of tumor-infiltrating FoxP3+ T cells in gastrointestinal cancers: a meta analysis. PLoS ONE 9(5): e94376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Hwang W-T, Adams SF, Tahirovic E, Hagemann IS, Coukos G (2012) Prognostic significance of tumor-infiltrating T-cells in ovarian cancer: a meta-analysis. Gynecol Oncol 124(2): 192–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Jochems C, Schlom J (2011) Tumor-infiltrating immune cells and prognosis: the potential link between conventional cancer therapy and immunity. Exp Biol Med 236(5): 567–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Kakavand H, Vilain RE, Wilmott JS, Burke H, Yearley JH, Thompson JF, Hersey P, Long GV, Scolyer RA (2015) Tumor PD-L1 expression, immune cell correlates and PD-1+ lymphocytes in sentinel lymph node melanoma metastases. Modern Pathol 28(12): 1535–1544. [DOI] [PubMed] [Google Scholar]
  40. Kang MJ, Kim KM, Bae JS, Park HS, Lee H, Chung MJ, Moon WS, Lee DG, Jang KY (2013) Tumor-infiltrating PD1-positive lymphocytes and FoxP3-positive regulatory T cells predict distant metastatic relapse and survival of clear cell renal cell carcinoma. Transl Oncol 6(3): 282–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Knief J, Reddemann K, Petrova E, Herhahn T, Wellner U, Thorns C (2016) High density of tumor-infiltrating B-lymphocytes and plasma cells signifies prolonged overall survival in adenocarcinoma of the esophagogastric junction. Anticancer Res 36(10): 5339–5345. [DOI] [PubMed] [Google Scholar]
  42. Knol AC, Nguyen JM, Quereux G, Brocard A, Khammari A, Dreno B (2011) Prognostic value of tumor-infiltrating Foxp3+ T-cell subpopulations in metastatic melanoma. Exp Dermatol 20(5): 430–434. [DOI] [PubMed] [Google Scholar]
  43. Kogashiwa Y, Yasuda M, Sakurai H, Nakahira M, Sano Y, Gonda K, Ikeda T, Inoue H, Kuba K, Oba S, Ishikawa J, Enoki Y, Matsumura S, Minami K, Ebihara Y, Sugasawa M (2017) PD-L1 expression confers better prognosis in locally advanced oral squamous cell carcinoma. Anticancer Res 37(3): 1417–1424. [DOI] [PubMed] [Google Scholar]
  44. Krpina K, Babarovic E, Jonjic N (2015) Correlation of tumor-infiltrating lymphocytes with bladder cancer recurrence in patients with solitary low-grade urothelial carcinoma. Virchows Archiv 467(4): 443–448. [DOI] [PubMed] [Google Scholar]
  45. Ladanyi A (2015) Prognostic and predictive significance of immune cells infiltrating cutaneous melanoma. Pigment Cell Melanoma Res 28(5): 490–500. [DOI] [PubMed] [Google Scholar]
  46. Ladanyi A, Kiss J, Mohos A, Somlai B, Liszkay G, Gilde K, Fejos Z, Gaudi I, Dobos J, Timar J (2011) Prognostic impact of B-cell density in cutaneous melanoma. Cancer Immunol Immunother 60(12): 1729–1738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Lee S-J, Lim HJ, Choi YH, Chang YH, Lee WJ, Kim DW, Yoon GS (2013) The clinical significance of tumor-infiltrating lymphocytes and microscopic satellites in acral melanoma in a Korean population. Ann Dermatol 25(1): 61–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Li JF, Chu YW, Wang GM, Zhu TY, Rong RM, Hou J, Xu M (2009) The prognostic value of peritumoral regulatory T cells and its correlation with intratumoral cyclooxygenase-2 expression in clear cell renal cell carcinoma. BJU Int 103(3): 399–405. [DOI] [PubMed] [Google Scholar]
  49. Liotta F, Gacci M, Frosali F, Querci V, Vittori G, Lapini A, Santarlasci V, Serni S, Cosmi L, Maggi L, Angeli R, Mazzinghi B, Romagnani P, Maggi E, Carini M, Romagnani S, Annunziato F (2011) Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU Int 107(9): 1500–1506. [DOI] [PubMed] [Google Scholar]
  50. Man Y-G, Stojadinovic A, Mason J, Avital I, Bilchik A, Bruecher B, Protic M, Nissan A, Izadjoo M, Zhang X, Jewett A (2013) Tumor-infiltrating immune cells promoting tumor invasion and metastasis: existing theories. J Cancer 4(1): 84–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Mao Y, Qu Q, Chen X, Huang O, Wu J, Shen K (2016) The prognostic value of tumor-infiltrating lymphocytes in breast cancer: a systematic review and meta-analysis. PLoS ONE 11(4): e0152500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Marvel D, Gabrilovich DI (2015) Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest 125(9): 3356–3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Mei Z, Liu Y, Liu C, Cui A, Liang Z, Wang G, Peng H, Cui L, Li C (2014) Tumour-infiltrating inflammation and prognosis in colorectal cancer: systematic review and meta-analysis. Br J Cancer 110(6): 1595–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Messaoudene M, Perier A, Fregni G, Neves E, Zitvogel L, Cremer I, Chanal J, Sastre-Garau X, Deschamps L, Marinho E, Larousserie F, Maubec E, Avril MF, Caignard A (2015) Characterization of the microenvironment in positive and negative sentinel lymph nodes from melanoma patients. PLoS One 10(7): e0133363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Mihm MC Jr., Mule JJ (2015) Reflections on the histopathology of tumor-infiltrating lymphocytes in melanoma and the host immune response. Cancer Immunol Res 3(8): 827–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Mittal D, Gubin MM, Schreiber RD, Smyth MJ (2014) New insights into cancer immunoediting and its three component phases—elimination, equilibrium and escape. Curr Opin Immunol 27: 16–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, Spector M, Perim D, Prince M, McLean S, Bradford CR, Taylor JM, Wolf GT (2016) Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck 38(7): 1074–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Nordfors C, Grun N, Tertipis N, Ahrlund-Richter A, Haeggblom L, Sivars L, Du J, Nyberg T, Marklund L, Munck-Wikland E, Nasman A, Ramqvist T, Dalianis T (2013) CD8+ and CD4+ tumour infiltrating lymphocytes in relation to human papillomavirus status and clinical outcome in tonsillar and base of tongue squamous cell carcinoma. Eur J Cancer 49(11): 2522–2530. [DOI] [PubMed] [Google Scholar]
  59. Obeid JM, Erdag G, Smolkin ME, Deacon DH, Patterson JW, Chen L, Bullock TN, Slingluff CL (2016) PD-L1, PD-L2 and PD-1 expression in metastatic melanoma: correlation with tumor-infiltrating immune cells and clinical outcome. Oncoimmunology 5(11): e1235107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Ozgur HH, Ercetin AP, Eliyatkin N, Seren A, Kupelioglu A, Ortac R, Diniz G, Aktas S (2014) Regulatory T cells and their prognostic value in hepatopancreatobiliary tumours. Hepatogastroenterology 61(135): 1847–1851. [PubMed] [Google Scholar]
  61. Park CK, Kim SK (2017) Clinicopathological significance of intratumoral and peritumoral lymphocytes and lymphocyte score based on the histologic subtypes of cutaneous melanoma. Oncotarget 8(9): 14759–14769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Park JH, Roxburgh CS, McMillan DC (2014) Comment on ‘Tumour-infiltrating inflammation and prognosis in colorectal cancer: systematic review and meta-analysis’. Br J Cancer 111(12): 2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Partlova S, Boucek J, Kloudova K, Lukesova E, Zabrodsky M, Grega M, Fucikova J, Truxova I, Tachezy R, Spisek R, Fialova A (2015) Distinct patterns of intratumoral immune cell infiltrates in patients with HPV-associated compared to non-virally induced head and neck squamous cell carcinoma. Oncoimmunology 4(1): e965570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Pretscher D, Distel LV, Grabenbauer GG, Wittlinger M, Buettner M, Niedobitek G (2009) Distribution of immune cells in head and neck cancer: CD8+ T-cells and CD20+ B-cells in metastatic lymph nodes are associated with favourable outcome in patients with oro- and hypopharyngeal carcinoma. BMC Cancer 9: 292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Punt S, Dronkers EA, Welters MJ, Goedemans R, Koljenovic S, Bloemena E, Snijders PJ, Gorter A, van der Burg SH, Baatenburg de Jong RJ, Jordanova ES (2016) A beneficial tumor microenvironment in oropharyngeal squamous cell carcinoma is characterized by a high T cell and low IL-17(+) cell frequency. Cancer Immunol Immunother 65(4): 393–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Saldanha G, Flatman K, Teo KW, Bamford M (2017) A novel numerical scoring system for melanoma tumor-infiltrating lymphocytes has better prognostic value than standard scoring. Am J Surg Pathol 41(7): 906–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, Symmans WF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S (2015) The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 26(2): 259–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Santoiemma PP, Powell DJ Jr (2015) Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther 16(6): 807–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331: 1565–1570. [DOI] [PubMed] [Google Scholar]
  70. Shankaran V (2001) IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410: 1107–1111. [DOI] [PubMed] [Google Scholar]
  71. Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, Bajorin DF, Reuter VE, Herr H, Old LJ, Sato E (2007) CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci USA 104(10): 3967–3972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Sideras K, Biermann K, Verheij J, Takkenberg BR, Mancham S, Hansen BE, Schutz HM, de Man RA, Sprengers D, Buschow SI, Verseput MC, Boor PP, Pan Q, van Gulik TM, Terkivatan T, Ijzermans JN, Beuers UH, Sleijfer S, Bruno MJ, Kwekkeboom J (2017) PD-L1, Galectin-9 and CD8+ tumor-infiltrating lymphocytes are associated with survival in hepatocellular carcinoma. Oncoimmunology 6(2): e1273309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Sjodahl G, Lovgren K, Lauss M, Chebil G, Patschan O, Gudjonsson S, Mansson W, Ferno M, Leandersson K, Lindgren D, Liedberg F, Hoglund M (2014) Infiltration of CD3(+) and CD68(+) cells in bladder cancer is subtype specific and affects the outcome of patients with muscle-invasive tumors. Urol Oncol 32(6): 791–797. [DOI] [PubMed] [Google Scholar]
  74. Song H, Wu Y, Ren G, Guo W, Wang L (2015) Prognostic factors of oral mucosal melanoma: histopathological analysis in a retrospective cohort of 82 cases. Histopathology 67(4): 548–556. [DOI] [PubMed] [Google Scholar]
  75. Stoll G, Bindea G, Mlecnik B, Galon J, Zitvogel L, Kroemer G (2015) Meta-analysis of organ-specific differences in the structure of the immune infiltrate in major malignancies. Oncotarget 6(14): 11894–11909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Sun DS, Zhao MQ, Xia M, Li L, Jiang YH (2012) The correlation between tumor-infiltrating Foxp3+ regulatory T cells and cyclooxygenase-2 expression and their association with recurrence in resected head and neck cancers. Med Oncol 29(2): 707–713. [DOI] [PubMed] [Google Scholar]
  77. Sun C, Xu J, Song J, Liu C, Wang J, Weng C, Sun H, Wei H, Xiao W, Sun R, Tian Z (2015) The predictive value of centre tumour CD8(+) T cells in patients with hepatocellular carcinoma: comparison with Immunoscore. Oncotarget 6(34): 35602–35615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Thomas NE, Busam KJ, From L, Kricker A, Armstrong BK, Anton-Culver H, Gruber SB, Gallagher RP, Zanetti R, Rosso S, Dwyer T, Venn A, Kanetsky PA, Groben PA, Hao H, Orlow I, Reiner AS, Luo L, Paine S, Ollila DW, Wilcox H, Begg CB, Berwick M (2013) Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol 31(33): 4252–4259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Tu J-F, Ding Y-H, Ying X-H, Wu F-Z, Zhou X-M, Zhang D-K, Zou H, Ji J-S (2016) Regulatory T cells, especially ICOS+ FOXP3+ regulatory T cells, are increased in the hepatocellular carcinoma microenvironment and predict reduced survival. Sci Rep 6: 35056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Ugel S, De Sanctis F, Mandruzzato S, Bronte V (2015) Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest 125(9): 3365–3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Wang B, Wu S, Zeng H, Liu Z, Dong W, He W, Chen X, Dong X, Zheng L, Lin T, Huang J (2015) CD103+ tumor infiltrating lymphocytes predict a favorable prognosis in urothelial cell carcinoma of the bladder. J Urol 194(2): 556–562. [DOI] [PubMed] [Google Scholar]
  82. Wang Q, Luan W, Warren L, Fiel MI, Blank S, Kadri H, Mandeli J, Hiotis SP (2016) Prognostic role of immune cells in hepatitis B-associated hepatocellular carcinoma following surgical resection depends on their localization and tumor size. J Immunother 39(1): 36–44. [DOI] [PubMed] [Google Scholar]
  83. Wansom D, Light E, Thomas D, Worden F, Prince M, Urba S, Chepeha D, Kumar B, Cordell K, Eisbruch A, Taylor J, Moyer J, Bradford C, D’Silva N, Carey T, McHugh J, Wolf G (2012) Infiltrating lymphocytes and human papillomavirus-16—associated oropharyngeal cancer. Laryngoscope 122(1): 121–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Ward MJ, Thirdborough SM, Mellows T, Riley C, Harris S, Suchak K, Webb A, Hampton C, Patel NN, Randall CJ, Cox HJ, Jogai S, Primrose J, Piper K, Ottensmeier CH, King EV, Thomas GJ (2014) Tumour-infiltrating lymphocytes predict for outcome in HPV-positive oropharyngeal cancer. Br J Cancer 110(2): 489–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Weiss SA, Han SW, Lui K, Tchack J, Shapiro R, Berman R, Zhong J, Krogsgaard M, Osman I, Darvishian F (2016) Immunologic heterogeneity of tumor-infiltrating lymphocyte composition in primary melanoma. Hum Pathol 57: 116–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Wolf GT, Chepeha DB, Bellile E, Nguyen A, Thomas D, McHugh J (2015) Tumor infiltrating lymphocytes (TIL) and prognosis in oral cavity squamous carcinoma: a preliminary study. Oral Oncol 51(1): 90–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Xu Q, Wang C, Yuan X, Feng Z, Han Z (2017) Prognostic value of tumor-infiltrating lymphocytes for patients with head and neck squamous cell carcinoma. Transl Oncol 10(1): 10–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Zhang L (2003) Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348: 203–213. [DOI] [PubMed] [Google Scholar]
  89. Zhang Q, Hao C, Cheng G, Wang L, Wang X, Li C, Qiu J, Ding K (2015) High CD4(+) T cell density is associated with poor prognosis in patients with non-muscle-invasive bladder cancer. Int J Clin Exp Pathol 8(9): 11510–11516. [PMC free article] [PubMed] [Google Scholar]
  90. Zhao HQ, Li WM, Lu ZQ, Yao YM (2014) Roles of Tregs in development of hepatocellular carcinoma: a meta-analysis. World J Gastroenterol 20(24): 7971–7978. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Cancer are provided here courtesy of Cancer Research UK

RESOURCES