Next Article in Journal
Current Molecular Profile of Gastrointestinal Stromal Tumors and Systemic Therapeutic Implications
Next Article in Special Issue
A Retrospective, Single-Institution Experience of Bullous Pemphigoid as an Adverse Effect of Immune Checkpoint Inhibitors
Previous Article in Journal
Antiplatelet Drugs on the Recurrence of Hepatocellular Carcinoma after Liver Transplantation
Previous Article in Special Issue
Checkpoint Inhibitors in Cancer Therapy: Clinical Benefits for Head and Neck Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints

by
Delphine Hudry
1,2,*,
Solenn Le Guellec
2,
Samuel Meignan
3,4,
Stéphanie Bécourt
2,
Camille Pasquesoone
2,
Houssein El Hajj
2,
Carlos Martínez-Gómez
2,
Éric Leblanc
1,2,
Fabrice Narducci
1,2 and
Sylvain Ladoire
5,6
1
Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
2
Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
3
Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
4
CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille University, F-59000 Lille, France
5
Department of Medical Oncology, Centre Georges-François Leclerc, F-21000 Dijon, France
6
INSERM, CRI-866 Faculty of Medicine, F-21000 Dijon, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(21), 5332; https://doi.org/10.3390/cancers14215332
Submission received: 6 September 2022 / Revised: 24 October 2022 / Accepted: 26 October 2022 / Published: 29 October 2022

Abstract

:

Simple Summary

Outcomes of ovarian cancer (OC) patients remain poor despite recent advances in oncology. Immunotherapy has proven to be beneficial in treating selected populations with different cancer types. However, OC shows very little response to immunotherapy; thus, it is mandatory to understand which subgroups of OC patients might benefit the most and how to potentiate its effect. In recent years, a more comprehensive understanding of the immune microenvironment of OC has been described, especially regarding the characterization of tumor-infiltrating lymphocytes (TILs). These results are promising and open the fields to new therapeutic approaches incorporating immune checkpoint blockade. This review aims to synthesize recent research on TILs in OC patients.

Abstract

Epithelial ovarian cancers (EOC) are often diagnosed at an advanced stage with carcinomatosis and a poor prognosis. First-line treatment is based on a chemotherapy regimen combining a platinum-based drug and a taxane-based drug along with surgery. More than half of the patients will have concern about a recurrence. To improve the outcomes, new therapeutics are needed, and diverse strategies, such as immunotherapy, are currently being tested in EOC. To better understand the global immune contexture in EOC, several studies have been performed to decipher the landscape of tumor-infiltrating lymphocytes (TILs). CD8+ TILs are usually considered effective antitumor immune effectors that immune checkpoint inhibitors can potentially activate to reject tumor cells. To synthesize the knowledge of TILs in EOC, we conducted a review of studies published in MEDLINE or EMBASE in the last 10 years according to the PRISMA guidelines. The description and role of TILs in EOC prognosis are reviewed from the published data. The links between TILs, DNA repair deficiency, and ICs have been studied. Finally, this review describes the role of TILs in future immunotherapy for EOC.

1. Introduction

Despite advances in medical treatment and surgery, ovarian cancer is the most lethal gynecological cancer. According to recent epidemiological estimates, there were 21,410 news cases of ovarian cancers in the United States in 2021, accounting for 13,770 deaths; this makes it the sixth most common cause of cancer-related death in women after lung, breast, colon, rectum, and pancreas cancers [1], with a 5-year survival rate of 46% [2] when all stages are combined. The most common histologic type is epithelial ovarian cancer (EOC), with 75% of the patients being diagnosed at advanced FIGO stages (III or IV) [2]. EOC includes a wide range of diseases with varying prognoses. The most common histologic subtypes are the following: high-grade serous ovarian carcinoma (HGSOC), which is usually associated with a poor prognosis; low-grade serous carcinoma (LGSOC); mucinous carcinoma; endometroid carcinoma; and clear cell carcinoma (CCC) [3]. These different subtypes present distinct anatomical origins, molecular profiles, and prognoses. The recommended first-line therapy includes complete cytoreduction associated with carboplatin–paclitaxel intravenous chemotherapy [4]. Three randomized controlled trials [5,6,7] showed that, in patients with a high tumor burden who are eligible for primary cytoreduction surgery (pCRS), neoadjuvant chemotherapy followed by interval cytoreduction surgery (iCRS) is associated with lower surgery-related morbidity. However, retrospective studies highlight the contrast between pCRS and iCRS patients’ profiles [8]. Patients who undergo iCRS usually present a high tumor burden that prevents upfront surgery. Patient evaluation with imaging and diagnostic laparoscopy is essential to determine the patient’s eligibility for pCRS [9]. The standard chemotherapy regimen consists of three-week cycles of carboplatin at AUC 5/6 and paclitaxel at 175 mg/m2 [10]. The number of cycles is determined by the tumor load, the disease stage, the presence of metastases, and the patient’s condition. In recent years, new treatment options, such as hyperthermic intraperitoneal chemotherapy (HIPEC) following iCRS [11,12], have been shown to improve overall survival (OS) and disease-free survival (DFS). Currently, studies are evaluating the impact of HIPEC during pCRS [13,14]. Adding bevacizumab, an antiangiogenic drug (vascular endothelial growth factor (VEGF) inhibitor), to chemotherapy or using it as maintenance therapy has been shown to result in an improvement in DFS in women with a high risk of disease progression [15,16]. Poly(-adenosine diphosphate ribose) polymerase (PARP) inhibitors have also been shown to have a beneficial effect when used in maintenance therapy [15,17]. The use of olaparib in maintenance therapy for HGSOC and endometrioid ovarian cancer with BRCA mutation decreases the risk of recurrence [18]. Since 2011, targeted immunotherapy drugs have opened new horizons for cancer treatment [19]. Immune checkpoint inhibitors (ICIs) are being developed and evaluated in various types of cancers [20]. The use of immunotherapy requires a better understanding of the different aspects of the tumor’s immune microenvironment. Recently, tumor immune infiltrates have been widely studied in EOC, particularly tumor-infiltrating lymphocytes (TILs).
This study aims to provide an overview of the scientific literature on TILs in EOC. TILs evaluation, prognostic impact, and relation with the new treatment options, PARP inhibitors and ICIs, are summarized in a systematic review.

2. Materials and Methods

This systematic literature review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses PRISMA guidelines [21], using specific eligibility criteria. We reviewed all articles identified through MEDLINE (via OVID) and EMBASE between January 2010 and August 2021. The combinations of mesh terms related to TIL subsets and EOC used in the search strategy are available in File S1. We included original English articles related to research studies using human EOC tissue. We excluded reviews, letters, and editorials. Studies focusing exclusively on tumor-associated neutrophils, macrophages, myeloid-derived suppressor cells, and natural killer cells were also excluded. The outcomes analyzed were TIL description, survival impact, and TIL modification with standard or new treatment.
All articles were independently screened for eligibility by two authors (DH and SLG). The process of selection was performed in two steps: the first step was the selection of the articles based on the titles and the abstracts, and the second step consisted of selecting the articles based on an evaluation of the full text. Two reviewers extracted the data independently and any disagreements were discussed with a third reviewer. For the included studies, data on study design, study period, number of patients, sample used, method and cut-off used to identify TILs, and other outcomes were extracted.

3. Results

3.1. Study Selection

Through our systematic search, 1279 unique articles were identified, including 211 that underwent full-text evaluation. The results of the review process are presented in Figure 1. After excluding articles with a too high risk of bias, study population, follow-up, and measurement of outcomes were examined. Ultimately, 122 original studies were included in the final review. The articles were grouped according to the main outcomes.

3.2. TIL Definition

  • TILs in EOC are a subject that has gained a lot of interest in the last five years. In the published scientific data, TILs are evaluated using different methods, including genetic signature, count of TILs in hematoxylin and eosin (H&E) based pathological immunohistochemistry (IHC), and immunofluorescence (IF) [22]. The International Immuno-Oncology Biomarkers Working Group defined, in 2017, “intra-epithelial” (iTILs) as TILs present in the tumor and “stromal” (sTILs) as TILs that are present within 1 mm beneath the epithelial layer [23]. To evaluate the inflammatory infiltrate, sTILs and iTILs are expressed in percentages or median counts. In IHC, between three and 10 fields in stained slides are observed in x200 or x400 high-power fields (HPF). TILs in H&E, in IHC, or in IF with specific antibodies are analyzed with either an absolute count or a semi-quantitative cut-off. For example, Goode and al. divided iTILs CD8+ infiltration into four categories: 0, low: 1–2, median: 3–19, and high ≥ 20 [24]. Other authors concluded that TILs > 5 or 10 per HPF should define positive iTILs in EOC [25].
  • Tumoral tissues and ascites from pCRS patients represent the main samples used in the studies [26]. The data showed variability in the immune infiltrate among the different tissue samples within the same patient (ovaries, omentum, and peritoneum) [27,28,29,30,31]. One study confirmed the feasibility of TIL evaluation in tumor samples that is performed using a 16-gauge needle biopsy [32]. Systematic tumor core biopsies can represent the immune microenvironment [33].
  • The description of TILs in EOC uses various techniques, especially the cut-offs are extremely variable between the studies. To date, there is no consensus, apart from H&E which is not yet used in clinical routine, on the type of marker or the thresholds to identify TILs.

3.3. TIL Phenotypes

In H&E, TIL evaluation includes mononuclear infiltrate, lymphocytes, and plasma cells [23]. Sometimes TILs are spotted using CD3 marking [34]. Various subsets of T cells may be found in EOC: CD8+ T cytotoxic and CD4+ T helper lymphocytes are identified by either the molecules on their surface or the pattern of cytokines they produce. T helper CD4+ cells are divided into subtypes: Th1 cells that produce interleukin 2 (IL-2) and interferon INF-𝛾 (acting on CD8+ cells); Th2 cells that produce IL-4, 5, 6, 10, and 13 (humoral immunity) [35]; Th17 cells that produce IL-17; and T Follicular helper (TFH) that interacts with B lymphocytes [36]. Tumor-infiltrating B lymphocytes (B-TILs) have been shown to be present in several solid tumors, including EOC [37]. Regulatory T cells (TREG) produce cytokines with immunosuppressive activities, including IL-10 and TGFβ [36]. The expressions of FOXP3 and CD25 often identify TREG. All immune cells are detected in different locations within and around the tumor epithelium. EOC is a heterogeneous disease regarding TILs. In terms of histologic subtypes, HGSOC is studied in the majority of studies. LGSOC, mucinous carcinoma, endometrioid carcinoma, and CCC are also studied in most studies, while carcinosarcoma is studied in only a few cases. Table 1 shows the description of TILs in the articles reviewed here. Only recent studies with relevant samples and/or results are shown to improve readability. In those studies, the number of cases ranges from a few dozen patients to several hundred, depending on the method used. The tumor stages studied vary, with most cases being at the most advanced stages (III and IV). Most of the materials used are derived from formalin-fixed, paraffin-embedded (FFPE) samples, with partial analysis (IHC, tissue microarray (TMA)) or global analysis (whole tissue sections (WTS), flow cytometry, gene expression profiles, and mRNA profiles). IHC sheds light on the heterogeneity between sTILs and iTILs [38]. A study of 37 cases of advanced EOC showed a good correlation between TMA and WTS regarding CD8+ TIL infiltration assessment [39]. CD3+TILs or sTILs [34] and Th17 [40,41] are present at a higher level in EOC than in borderline or benign ovarian. Th1, Th2, and Th17 profiles are diverse within the same patient between tumor and ascites or intra-cystic fluid [29,40,42,43], and between omentum and ovarian tissue [44]. The absolute median count [45] and the CD4/CD8 ratio have a high variability [46]. CD3+T-cells in the ascites increase with a higher frequency of CD4+CD45RA-FoxP3+ T-cells in the ascites compared to the peripheral blood [47]. The frequency of TREG increases in the ovarian tumors compared to the blood samples [48]. Heterogeneity is also seen between tumor grades, with TREG and CD8+ TILs being higher in grade 2 or 3 than in grade 1 [49]. Tertiary lymphoid structures (TLS) are present in HGSOC [50], with a potential role in immunosuppression. Figure 2A is a simplified view of the immunologic network at the tumor site.
A new way to classify cancers is based on gene expression profiles. Using this way, in 2011, the Cancer Genome Atlas (TCGA) divided HGSOC into four distinct groups: mesenchymal, immunoreactive, proliferative, and differentiated [51]. The most used algorithms for genetic analysis, ESTIMATE [52] or CIBERSORT [53], have been used in 379 cases of EOC [54], with 22 immune cells being studied, and confirmed the wide variability in TILs between tumors, especially in CD8+ or TFH. Using the same method in a large EOC cohort (n = 2086, stages I to IV), TCGA and CIBERSORT algorithms allowed us to underline the heterogeneity and to highlight a proportion of 12.2% of macrophages, 6.6% of TFH, and 6.3% of memory CD4 T cells among the TILs [55]. TCGA analysis of 3176 EOC samples illustrate, as expected, the heterogeneity between histologic subtypes, especially between high- and low-grade serous carcinomas [56]. Murakami established an IHC classification that distinguishes four subgroups: mesenchymal transition, immune reactive, solid and proliferative, and papilloglandular [57]. The new classification of IHC has been used in 70 ovarian or peritoneal samples and confirms the heterogeneity in sTIL density [58]. In particular, the heterogeneity in TILs is observable in the primary tumor versus recurrence [59]. For example, the median FOXP3 count is higher during recurrence than at diagnosis [60]. Thus, new techniques are used, including spatioimageomic transcriptomics [61] and imaging mass cytometry +/− combined with machine learning approaches [62], to specify the phenotypic and spatial heterogeneities in TILs in EOC.
The specific markers provide the knowledge on TILs infiltration in EOC, including the type of T cells, cytotoxic T cells, and suppressors. The clinical cohorts used only partially reflect the disease. Indeed, most data are analyzed based on operable diseases from diagnosis, which unfortunately constitute only a part of the patients in practice.
Table 1. TIL phenotypes in the reviewed studies.
Table 1. TIL phenotypes in the reviewed studies.
Study Author (Publication Year)Number of Cases n=Tumor StageMomentSubtype/ TIL PhenotypeSpecimen ProcessingLocationTIL Description
Hagemann (2011) [63]10IIICpCRS (n = 9) and recurrence (n = 1)CD3, CD8, FoxP3primary tumor and two intraperitoneal metastasesiTILs and sTILsheterogeneity in TIL density inter- and intra-patient (primary versus metastasis)
Murakami (2016) [57]132I to IVat diagnosis: pCRSCD8tumor samplesiTILs and sTILsNEW PATHOLOGICAL CLASSIFICATION: mesenchymal transition, immunereactive, solid and proliferative, and papilloglandular
Ojalvo (2017) [64]52II to IVpCRS (37) and recurrence (15)CD8, FoxP3tumor samplesiTILsmedian FOXP3 count recurrent > primary
Zhu (2017) [41]126I to IVpCRSCD4, IL17, FoxP3, CD31ovarian samples of the central areas of EOCiTILs% of Treg cells, Th17 cells, and ratio of Treg/Th17 cells: high in patients with EOC
Nakamura (2019) [65]839no datano dataTh1, Th2, Th9, Th17, M1, and M2 macrophagetumor samplesTILsHigher intratumoral expressionmarkers may rescue or neutralize the negative associations of inflammation or angiogenesis
Jiménez (2020) [27]50IIIC and IV40 NACT and 10 pCRSCD45, CD3, CD4, CD8, NK, FoxP3tumor samplesiTILstranscriptomic heterogeneity in each patient
Dötzer (2019) [30]49IIIC and IVpCRS
(35 CC0, 8 CC1, 6 CC2)
CD45, CD3, CD8, PD-1, PD-L1tumor sample, peritoneum, and omentumiTILs and sTILsdifferences in the expression between primary cancer and omental and peritoneal lesions
Oberg (2020) [43]29III and IVat diagnosisCD45, CD8, CD56, CD3, IFN-𝛾, IL-4, IL-9, IL-10, IL-17, TNF-𝛼blood, ascites, and tumor samplesTILsheterogeneity in TILs vs ascites
Gao (2020) [55]2086I to IVno data22 immune cellstumor samplesTILsHeterogeneous immune microenvironment: infiltration varied between clinicopathological subgroups (stage, type, and survival)
Lakis (2020) [58]70III and IVpCRSH&E, morphological subtypes: IR, SD, PG, MTtumor samplesiTILs and sTILshigher sTIL density in implants than in ovarian tissue; heterogeneity between implants of the same patient
Zhou (2021) [54]379II to IVno data22 immune cellstumor samplesTILsheterogeneity in CD8 and NK cells; TFH, monocyte, macrophage: proportion higher in OC than normal tissue
Zhu (2021) [62]41IIIB and IVpCRSCD8+, CD4+tumor samplesiTILs and sTILsheterogeneity between stromal and tumoral tissues
Karakaya (2021) [38]66I to IVat diagnosisPD-1, CD8, CD4, CD3tumor samplesiTILs and sTILsheterogeneity between stromal and tumoral tissues and between histologic types
This table summarizes the studies describing TILs in EOC. The description of the clinical cohort (number of cases, tumor stage, and moment), the type of T cells studied, the tissue specimen, and the location (stroma or intraepithelial) is correlated with the main outcome about TIL description for each of the articles. pCRS: primary cytoreductive surgery, iCRS: intervalle cytoreductive surgery, iTILs: intra-tumoral TILs, sTILs: stromal TILs, NACT: neoadjuvant chemotherapy, OC: ovarian cancer, vs: versus, IR: immune reactive, SD: solid and proliferative, PG: papilloglandular, MT: mesenchymal transition.

3.4. TILs and Patients’ Survival

The survival benefits of TILs in EOC have been noted for a long time. In 2003, Coukos and colleagues analyzed 186 advanced EOC tissue samples and detected intra-epithelial CD3+ (iCD3) TILs in 55% of the patients. The 5-year survival rate of these patients was 38%, compared to 4.5% in patients with no detectable TILs [66]. Figure 3 summarizes the studies published in the last ten years that evaluated the impact of TILs on survival. Table 2 shows the characteristics of the patients in the different studies. In the absence of specific antibodies in IHC, most studies employed the H&E analysis [57,67,68,69] with median count or semi-quantitative categories, and survival rates were evaluated (OS and DFS). OS is calculated from the date of histological diagnosis to death (or, in rare cases, from the date of first treatment to the date of death), and DFS is calculated from the date of beginning of treatment to the date of progression or death.
In 2016, Murakami et al. confirmed the positive effect on OS and DFS; the immunoreactive subtype is the group with the better prognosis compared to three of the other groups [57]. TILs are known to be associated with favorable prognostic factors in many solid tumors, including HGSOC [25]. Different types of infiltrating immune cells have varying effects on the prognosis of the patients [36]. CD8+ TILs [66,70,71], Th1 TILs, and Th17 TILs [65,72] are associated with a positive effect. Th2 TILs are associated with either a negative [29,56] or a positive impact [65]. TREG TILs are associated with either a negative [73] or a positive impact [74,75]. CD3-staining iTILs based on the TILs count are counted either manually or using digital imaging analysis to determine the number of T cells per HPF (ranging from 15 to 20 HPF). Several studies found a positive effect of CD3+ TILs [44,63,76,77,78,79], and one showed no impact on the prognosis [80]. The CD3 location can modify the prognosis: sCD3+ TILs are associated with an improved 10-year survival rate [81], whereas iCD3+ TILs have no impact on OS or DFS [82]. Moreover, the studies’ results are sometimes contrasting, with an effect on DFS but not on OS [83], or with a positive effect on OS only [84]. In one study, at diagnosis, most patients showed stromal CD3+ immune infiltration with high heterogeneity in the intra-epithelial CD3+ [85]. In this cohort, while an increase in stromal CD8+/FoxP3+ over 10-fold was associated with a better OS, no association was observed when considering iTILs counts [85].
CD4+ memory TILs (CD45RO+) are most frequently positively correlated with DFS or OS [56,79,86]. This is the case for iCD4+ TILs, but not sCD4+ TILs [87]. However, this positive impact is not consistent [80,82,88]. A significant infiltration of CD8 TILs is most often associated with a positive prognostic effect on OS [24,54,56,76,77,79,81,89,90,91,92]. When only intraepithelial CD8 cells are studied, the results are more heterogeneous, with one study having a positive effect on OS [24], two studies having a positive effect on DFS [87,93], three studies without a significant impact [39,94,95], and three studies having a negative effect on survival [82,96,97]. The results for TREG also vary, with either studies showing a negative impact on OS and DFS [45,78,90,94], or finding no impact or [49,79,82] even a positive impact [45,98].
Some studies evaluated B-TILs and TILs and showed a positive impact on survival [50,81,88,89,99], while others showed no impact at all [82,100]. Using machine learning-based refined differential gene expression and marker combination analysis, 44 markers were evaluated in 839 patients. A higher expression of Th1, Th2, and Th17 associated genes was correlated with better survival outcomes [65]. A 2086 SOC cohort showed that TFH had a negative impact on prognosis [55]. In HGSOC, the presence of CD103 with CD3+ and CD4+ was correlated with a better survival [97,101].
Altogether, recent studies are more controversial than the original study. The variability in the results can be explained by OC heterogeneity, different histology, and intra-patient variability. The clinical cohorts used are also an explanation. Indeed, iCRS are not represented in recent studies (Table 2), and the complete surgery rate is unfrequently used in favor of the optimal surgery rate, which is sometimes much lower than the recommendations for good practice. These confounding factors, type of sample used, and homogeneity of the clinical cohorts reduce the comparability between the studies.
Table 2. Patient characteristics in studies evaluating prognostic impact.
Table 2. Patient characteristics in studies evaluating prognostic impact.
Study Author
(Publication Year)
Number of Cases
n=
Advanced Stage
(%)
Serous Histology
(%)
MomentMedian Follow-Up (Months)Optimal Debulking (Residual Tumor <2.5 cm) (%)
Nielsen (2012) [89]26492.5100pCRS1.930
Bachmayr-Heyda (2013) [93]20395.588.2pCRS4869.5
Webb (2014) [101]49758.944.3pCRSno data100
Hermans (2014) [90]21010077.1At diagnosisno data30
De Leeuw (2015) [91]18734100pCRSno data100
Murakami (2016) [57]13282100pCRSno datano data
Santoiemma (2016) [81]1357165.9no datano data59.3
Lundgren (2016) [100]154no data58.4pCRS87no data
Goode (2017) [24]507847.862.9pCRS48.942.4
James (2017) [67]7074044.4pCRSno datano data
Pinto (2018) [87]12880.5100At diagnosisno data80
Hwang (2019) [68]2566256.6At diagnosisno data91
Martin de la Fuente (2020) [84]130100100At diagnosisno data60
Paijens (2021) [97]26894.8100pCRS (47%) or iCRSno data75.7 (51.4 complete)
Wu (2021) [102]44187no datano datano data62.4
Li (2021) [103]30888.9no datano datano data61.4 (18.2 complete)
Chen (2020) [104]18959.312.7At diagnosis37no data
This table details the clinical characteristics in studies evaluating the prognostic impact of TILs in EOC. Management of EOC is pCRS or iCRS when primary surgery is not possible. Optimally, the goal of surgery is complete resection, with residual tumor: 0 mm. pCRS: primary cytoreductive surgery, iCRS: intervalle cytoreductive surgery, and NACT: neoadjuvant chemotherapy.

3.5. Influence of DNA Repair Deficiency and TILs

Genetic alterations in ovarian cancer are dependent on diverse genes. Mismatch repair (MMR) pathway dysregulation represents 5 to 13 % [105] of SOC (including Lynch Syndrome), and BRCA mutations and alterations in homologous recombination are present in 23% and 50%, respectively [106]. Dysregulation of genes implicated in DNA repair leads to a higher mutational burden in ovarian cancers [107], and patients with homologous recombination deficiency (HRD) have been reported to have a higher expression of neoantigens [108,109]. Wang et al. reported a study using TCGA database to analyze the infiltration pattern in ovarian cancer. They classified the tumors into two clusters. The cluster enriched in cytotoxic and immunosuppressive cells tended to have a higher mutational load than the cluster with less immune cell infiltration [110]. Likewise, several studies showed a significantly higher number of CD3+ and CD8+ TILs in HRD [109,111,112] and microsatellite instability tumors [105,113]. There seems to be no difference in TIL infiltrations rates between BRCA1- and BRCA2-mutated patients [109,114]. However, it is noteworthy that homologous recombination proficient (HRP) patients are a heterogeneous group. In this group, some patients express high HLA class 1 molecule and high expression of neoantigens; thus, the accumulation of DNA mutations is not the only process implicated in patients with high immune infiltrate [108]. Understandably, there is more programmed death-ligand (PD-L)1 expression in HRD+ tumors than in HRP when considering combined positive score (CPS); however, interestingly, PD-L1 expression in tumoral cells seems to be equal in the two groups [109]. To conclude, a hypothesis is that high neoantigen load leads to the recruitment of TILs, which is counterbalanced by the expression of immune checkpoints. Lastly, it has been reported that some ovarian cancers with DNA repair deficiency escape immune surveillance despite being a «hot» phenotype according to the morphological diversification of the tumors. This mechanism is being evaluated in 514 cases of advanced HGSOC using TCGA analysis [115]. The ESTIMATE and ABSOLUTE algorithm applications highlight the spatial heterogeneity [115]. Anti-PARPs provide new options for patients presenting with a HRD+ tumor. Understanding the TIL landscape for these tumors is very helpful to propose anti-PARPs in combination with immunotherapy.

3.6. TILs and Immune Checkpoints

Programmed death-1 (PD-1 or CD279), with the activation-induced expression on T-cells, can bind to its ligands PD-L1 and PD-L2 to decrease the ability of TILs in destroying tumor cells. PD-L1 overexpression is one way for ovarian cancer to escape the immune surveillance [113]. Monoclonal antibodies targeting the immune inhibitory checkpoints, such as PD-1 and PD-L1, have been tested to evaluate the intensity and quality of T-cell activation [116,117]. PD-(L)1 inhibitor has been approved in the treatment of melanoma, non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, urothelial carcinoma, renal cell cancer, and cervical cancer [118]. Biomarkers used to guide treatment are PD-L1s evaluated with IHC, tumor mutation burden, or mismatch repair. In the past, PD-L1 tumor staining was widely used to determine the responders in treated patients. Tumor proportion score (TPS) for PD-L1 is representative of the proportion of tumor cells with membranous PD-L1 expression. More recent studies incorporate the presence of PD-1 or PD-L1 in TILs [118]. Combined positive score (CPS) includes the number of tumor cells, lymphocytes, and macrophages stained with PD-L1 divided by the total number of viable tumor cells and then multiplied by 100 [119]. Figure 2B illustrates the main immune checkpoints studied in EOC. It has been developed to better predict the response to immunotherapy. Table 3 shows the scores in the main-reviewed studies. Scoring expression in the immune checkpoint molecules, the definition of positive TILs, and PD-L1 positive tumors are very heterogeneous. Various studies evaluating HGSOC tissue samples showed that high expressions of PD-1 and PD-L1 in tumors are associated with a better DFS [84,120,121,122,123]. Combined analysis of PD-L1 expression in tumors and CD8+ iTILs allows the stratification of patients based on their prognosis: patients with negative PD-L1 expression in tumors and higher numbers of CD8+ iTILs have the longest median OS, while those with positive PD-L1 expression in tumors and lower numbers of CD8+ iTILs have the shortest median OS [124]. The high density of sTILs-PD-L1+ is associated with a favorable prognostic effect on OS [125]. Exhausted status of CD8+ TILs can be assessed by the co-expression of PD-1 and Tim3 and has been linked to poor prognosis [126]. The localization of the studied tissues seems to have an impact on the prognostic value: TILs expressing PD-1 in carcinomatosis tissue are associated with a better OS and PD-L1 expression in peritoneal tissue is negatively correlated with OS [127]. The expressions of other TIL immune checkpoints in OC, such as T cell immunoglobulin, mucin domain-containing protein 3 (Tim-3), lymphocyte activating gene 3 (LAG-3), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4), have also been evaluated [123]. These checkpoints are also the different targets for other ICIs. Blocking CTLA4, for example, activates CD8+ and CD4+ T cells and enhances the anti-tumor effect of drugs [116]. Targeting Tim-3 is tested in association with anti PD-1 [128]. LAG3 expression has been found to be associated with PD-L1 expression (intra-tumor PD-L1 and CPS score ≥ 1) in 48 HGSOC patients [114], which could lead to combination therapy targeting PD-L1 and LAG3 together.
The contrast in the IC study results can be explained by sampling variation, differences in staining protocols, variability in cut-off values, and variability in the localization of tumors or TILs. TPS and CPS for PD-L1 scoring are approved by the Food and Drug Administration. Immune-checkpoint analysis, particularly TILs, is very likely to be integrated in the future to screen OC patients who might benefit from targeted therapy. A precise description of the immune checkpoints presents in the TILs and on the tumor cells is certainly useful in future trials of ICIs in EOC aiming to appropriately stratify the patients.

3.7. Influence of First-Line Chemotherapy on TIL Landscape

We reviewed above the prognostic significance of TILs reported by several studies, most of which were performed on patients undergoing pCRS, as shown in Table 2. Yet, most patients receive platinum-based neoadjuvant chemotherapy (NACT) before iCRS. Depending on the studies, the proportion of CD3+ cells before and after NACT could either be identical [129,130], decreased [131], or increased [69,85]. One explanation for these results is the high inter-patient and intra-patient variability [27]. A way to highlight this heterogeneity is to compare site-matched metastases before and after NACT. A transcriptomic study of 38 matched samples showed an oncogenic expression profile before NACT that evolved to an immune expression profile during chemotherapy. The results showed an increase in the proportion of NK cells and cytotoxic gene set expression after NACT, with no difference in the other subpopulations of TILs. T-cell receptor (TCR) sequencing showed expansion of oligoclonal TCR after NACT in site-matched samples. Other studies showed an increase in CD8+/CD4+ and CD8+/FoxP3+ ratios after NACT [85,132]. These results suggest an anti-tumoral response to chemotherapy with recruitment of NK cells, a decrease in regulatory cells, activation of cytotoxic response, and clonal expansion of T cells.
The prognostic significance of TIL evolution with NACT was explored in 54 patients with advanced ovarian cancer [131]: TIL subpopulations were studied before and after NACT in patients with a good Chemotherapy Response Score (CRS) compared to patients with a poor CRS [133]. Their results showed no differences in immune infiltration density between good and poor responders before NACT. After NACT, there was a decrease in sFoxP3+ cells in good responders, but no difference was seen in poor responders. Good responders showed an increase in IFN-𝛾 expression and a gene expression profile of Th1 activation. There was also an increase in PDL1 expression after NACT in both groups, as reported elsewhere [134]. The results also showed an anti-tumoral response to chemotherapy and a reaction of tumoral cells by expressing PDL1. Therefore, immunotherapy could be more efficient after the first line of chemotherapy.
Most studies involved sample analysis during pCRS. TILs variation that is compared between pre- and post-chemotherapy differs among the studies. It has already been shown that chemotherapy has the potential to alter immunotherapy response [135,136]. The proportion of CD3 TILs does not change after NACT [129,130], whereas NACT induces a decrease in the density of sCD3 TILs in HGSOC patients [131], or an increase in the proportions of sCD3, sCD8, and iCD8 TILs [69,137]. sTILs are associated with platinum sensitivity in 70 patients with advanced-stage SOC [58]. Chemotherapy induces an upregulation of PD-L1 [69,130]. In recurrent HGSOC, a higher density of TILs and a higher expression of MHC have been reported when compared to paired primary tumors, suggesting a higher immunogenicity [85]. The repertoire of neo-epitope recognizing T-cells and their avidity are also different between blood and tumor samples in recurrent disease [138]. The study of TIL evolution, from state of activation to neo-epitope repertoire, under chemotherapy pressure could be key to the development of new personalized immunotherapy.

4. Conclusions

Even though immunotherapy is less efficient in EOC compared to other solid tumors, ongoing trials are evaluating the efficacy of combining standard treatments with immunotherapy to improve patients’ prognosis [139,140]. Treatment options that are being evaluated include associating TILs with chemotherapy, antiangiogenic drugs, PARP inhibitors, vaccines, cytokine injections, CAR-T cells, checkpoint inhibitors [141], and even radiotherapy [139,142]. Indeed, combinations of anti-PARP treatment and reactivation of the immune system via anti-PD-L1, PD-L2, or CTLA4 are among the therapeutic options tested in EOC. Standardizing TIL evaluation methods, techniques, and cut-offs is mandatory and is being evaluated in ongoing studies. The biggest challenge now is to harmonize TIL count and immune checkpoint scoring to help develop care strategy. Future personalized medicine will most certainly use the description of the tumor microenvironment, including TILs.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cancers14215332/s1, File S1: search strategy.

Author Contributions

Conceptualization, D.H. and S.L.; methodology, D.H. and S.L.; software, D.H.; validation S.M., S.B., É.L., and S.L.; formal analysis, D.H. and S.L.G.; investigation, D.H., S.B., and É.L.; resources, F.N.; data curation, D.H.; Writing—original draft preparation, D.H. and S.L.G.; Writing—review and editing, S.M., S.B., C.P., H.E.H., C.M.-G., É.L., F.N., and S.L.; visualization S.M., S.B., C.P., H.E.H., C.M.-G., É.L., F.N., and S.L.; Supervision, S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank Séverine Marchant for technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Lheureux, S.; Gourley, C.; Vergote, I.; Oza, A.M. Epithelial ovarian cancer. Lancet 2019, 393, 1240–1253. [Google Scholar] [CrossRef] [Green Version]
  3. World Health Organization (WHO). Classification of Tumours Editorial Board. Female Genital Tumours; World Health Organization Classification of Tumours: Lyon, France, 2020; Volume 4. [Google Scholar]
  4. Armstrong, D.K.; Alvarez, R.D.; Bakkum-Gamez, J.N.; Barroilhet, L.; Behbakht, K.; Berchuck, A.; Berek, J.S.; Chen, L.M.; Cristea, M.; DeRosa, M.; et al. NCCN Guidelines Insights: Ovarian Cancer, Version 1.2019. J. Natl. Compr. Cancer Netw. 2019, 17, 896–909. [Google Scholar] [CrossRef] [Green Version]
  5. Vergote, I.; Tropé, C.G.; Amant, F.; Kristensen, G.B.; Ehlen, T.; Johnson, N.; Verheijen, R.H.; van der Burg, M.E.; Lacave, A.J.; Panici, P.B.; et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 2010, 363, 943–953. [Google Scholar] [CrossRef] [Green Version]
  6. Kehoe, S.; Hook, J.; Nankivell, M.; Jayson, G.C.; Kitchener, H.; Lopes, T.; Luesley, D.; Perren, T.; Bannoo, S.; Mascarenhas, M.; et al. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): An open-label, randomised, controlled, non-inferiority trial. Lancet 2015, 386, 249–257. [Google Scholar] [CrossRef]
  7. Fagotti, A.; Ferrandina, M.G.; Vizzielli, G.; Pasciuto, T.; Fanfani, F.; Gallotta, V.; Margariti, P.A.; Chiantera, V.; Costantini, B.; Gueli Alletti, S.; et al. Randomized trial of primary debulking surgery versus neoadjuvant chemotherapy for advanced epithelial ovarian cancer (SCORPION-NCT01461850). Int. J. Gynecol. Cancer 2020, 30, 1657–1664. [Google Scholar] [CrossRef]
  8. Kraus, F.; El Hajj, H.; Le Deley, M.-C.; Aissaoui, O.; Gachon, B.; Chevalier, A.; Abdeddaim, C.; Lemaire, A.-S.; Ben Haj Amor, M.; Sylla, D.; et al. A New Paradigm in Managing Advanced Ovarian Cancer: Differentiating Patients Requiring Neoadjuvant Treatment from Primary Cytoreduction. Cancers 2021, 13, 4925. [Google Scholar] [CrossRef]
  9. Straubhar, A.M.; Filippova, O.T.; Cowan, R.A.; Lakhman, Y.; Sarasohn, D.M.; Nikolovski, I.; Torrisi, J.M.; Ma, W.; Abu-Rustum, N.R.; Gardner, G.J.; et al. A multimodality triage algorithm to improve cytoreductive outcomes in patients undergoing primary debulking surgery for advanced ovarian cancer: A Memorial Sloan Kettering Cancer Center team ovary initiative. Gynecologic oncology 2020, 158, 608–613. [Google Scholar] [CrossRef]
  10. Blagden, S.P.; Cook, A.D.; Poole, C.; Howells, L.; McNeish, I.A.; Dean, A.; Kim, J.W.; O’Donnell, D.M.; Hook, J.; James, E.C.; et al. Weekly platinum-based chemotherapy versus 3-weekly platinum-based chemotherapy for newly diagnosed ovarian cancer (ICON8): Quality-of-life results of a phase 3, randomised, controlled trial. Lancet Oncol. 2020, 21, 969–977. [Google Scholar] [CrossRef]
  11. van Driel, W.J.; Koole, S.N.; Sikorska, K.; Schagen van Leeuwen, J.H.; Schreuder, H.W.R.; Hermans, R.H.M.; de Hingh, I.; van der Velden, J.; Arts, H.J.; Massuger, L.; et al. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. N. Engl. J. Med. 2018, 378, 230–240. [Google Scholar] [CrossRef]
  12. Lei, Z.; Wang, Y.; Wang, J.; Wang, K.; Tian, J.; Zhao, Y.; Chen, L.; Luo, J.; Jia, M.; Tang, H.; et al. Evaluation of Cytoreductive Surgery with or without Hyperthermic Intraperitoneal Chemotherapy for Stage III Epithelial Ovarian Cancer. JAMA Netw. Open 2020, 3, e2013940. [Google Scholar] [CrossRef]
  13. Koole, S.; van Stein, R.; Sikorska, K.; Barton, D.; Perrin, L.; Brennan, D.; Zivanovic, O.; Mosgaard, B.J.; Fagotti, A.; Colombo, P.E.; et al. Primary cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy (HIPEC) for FIGO stage III epithelial ovarian cancer: OVHIPEC-2, a phase III randomized clinical trial. Int. J. Gynecol. Cancer 2020, 30, 888–892. [Google Scholar] [CrossRef]
  14. El Hajj, H.; Vanseymortier, M.; Hudry, D.; Bogart, E.; Abdeddaim, C.; Leblanc, E.; Le Deley, M.C.; Narducci, F. Rationale and study design of the CHIPPI-1808 trial: A phase III randomized clinical trial evaluating hyperthermic intraperitoneal chemotherapy (HIPEC) for stage III ovarian cancer patients treated with primary or interval cytoreductive surgery. ESMO Open 2021, 6, 100098. [Google Scholar] [CrossRef]
  15. Ray-Coquard, I.; Pautier, P.; Pignata, S.; Perol, D.; Gonzalez-Martin, A.; Berger, R.; Fujiwara, K.; Vergote, I.; Colombo, N.; Maenpaa, J.; et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2416–2428. [Google Scholar] [CrossRef]
  16. Perren, T.J.; Swart, A.M.; Pfisterer, J.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; Kurzeder, C.; et al. A phase 3 trial of bevacizumab in ovarian cancer. N. Engl. J. Med. 2011, 365, 2484–2496. [Google Scholar] [CrossRef] [Green Version]
  17. Moore, K.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2018, 379, 2495–2505. [Google Scholar] [CrossRef]
  18. DiSilvestro, P.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Efficacy of Maintenance Olaparib for Patients with Newly Diagnosed Advanced Ovarian Cancer with a BRCA Mutation: Subgroup Analysis Findings From the SOLO1 Trial. J. Clin. Oncol. 2020, 38, 3528–3537. [Google Scholar] [CrossRef]
  19. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  20. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  21. Moher, D.; Shamseer, L.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; Group, P.-P. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 2015, 4, 1. [Google Scholar] [CrossRef]
  22. Hendry, S.; Salgado, R.; Gevaert, T.; Russell, P.A.; John, T.; Thapa, B.; Christie, M.; van de Vijver, K.; Estrada, M.V.; Gonzalez-Ericsson, P.I.; et al. Assessing Tumor-infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immunooncology Biomarkers Working Group: Part 1: Assessing the Host Immune Response, TILs in Invasive Breast Carcinoma and Ductal Carcinoma In Situ, Metastatic Tumor Deposits and Areas for Further Research. Adv. Anat. Pathol. 2017, 24, 235–251. [Google Scholar] [CrossRef] [Green Version]
  23. Hendry, S.; Salgado, R.; Gevaert, T.; Russell, P.A.; John, T.; Thapa, B.; Christie, M.; van de Vijver, K.; Estrada, M.V.; Gonzalez-Ericsson, P.I.; et al. Assessing Tumor-Infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non-Small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv. Anat. Pathol. 2017, 24, 311–335. [Google Scholar] [CrossRef]
  24. Goode, E.L.; Block, M.S.; Kalli, K.R.; Vierkant, R.A.; Chen, W.; Fogarty, Z.C.; Gentry-Maharaj, A.; Tołoczko, A.; Hein, A.; Bouligny, A.L.; et al. Dose-Response Association of CD8+ Tumor-Infiltrating Lymphocytes and Survival Time in High-Grade Serous Ovarian Cancer. JAMA Oncol. 2017, 3, e173290. [Google Scholar] [CrossRef] [Green Version]
  25. Hwang, W.T.; Adams, S.F.; Tahirovic, E.; Hagemann, I.S.; Coukos, G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: A meta-analysis. Gynecol. Oncol. 2012, 124, 192–198. [Google Scholar] [CrossRef] [Green Version]
  26. Jang, M.; Yew, P.Y.; Hasegawa, K.; Ikeda, Y.; Fujiwara, K.; Fleming, G.F.; Nakamura, Y.; Park, J.H. TCR profiling of T lymphocytes in ovarian tumors and malignant ascites using next-generation sequencing. Cancer Res. 2015, 75, 4887. [Google Scholar] [CrossRef]
  27. Jiménez-Sánchez, A.; Cybulska, P.; Mager, K.L.V.; Koplev, S.; Cast, O.; Couturier, D.L.; Memon, D.; Selenica, P.; Nikolovski, I.; Mazaheri, Y.; et al. Unraveling tumor–immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy. Nat. Genet. 2020, 52, 582–593. [Google Scholar] [CrossRef]
  28. Foord, E.; Arruda, L.C.M.; Gaballa, A.; Klynning, C.; Uhlin, M. Characterization of ascites- and tumor-infiltrating γδ T cells reveals distinct repertoires and a beneficial role in ovarian cancer. Sci. Transl. Med. 2021, 13, eabb0192. [Google Scholar] [CrossRef]
  29. Cândido, E.B.; Silva, L.M.; Carvalho, A.T.; Lamaita, R.M.; Filho, R.M.P.; Cota, B.D.C.V.; Da Silva-Filho, A.L. Immune response evaluation through determination of type 1, type 2, and type 17 patterns in patients with epithelial ovarian cancer. Reprod. Sci. 2013, 20, 828–837. [Google Scholar] [CrossRef]
  30. Dötzer, K.; Schlüter, F.; Bo Schoenberg, M.; Bazhin, A.V.; von Koch, F.E.; Schnelzer, A.; Anthuber, S.; Grab, D.; Czogalla, B.; Burges, A.; et al. Immune heterogeneity between primary tumors and corresponding metastatic lesions and response to platinum therapy in primary ovarian cancer. Cancers 2019, 11, 1250. [Google Scholar] [CrossRef] [Green Version]
  31. Tran, E.; Nielsen, J.S.; Wick, D.A.; Ng, A.V.; Johnson, L.D.S.; Nesslinger, N.J.; McMurtrie, E.; Webb, J.R.; Nelson, B.H. Polyfunctional t-cell responses are disrupted by the ovarian cancer ascites environment and only partially restored by clinically relevant cytokines. PLoS ONE 2010, 5, e15625. [Google Scholar] [CrossRef]
  32. Hagemann, A.R.; Cadungog, M.; Hagemann, I.S.; Hammond, R.; Adams, S.F.; Chu, C.S.; Rubin, S.C.; Zhang, L.; Addya, K.; Birrer, M.J.; et al. Tissue-based immune monitoring I: Tumor core needle biopsies allow in-depth interrogation of the tumor microenvironment. Cancer Biol. Ther. 2011, 12, 357–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lara, O.D.; Krishnan, S.; Wang, Z.; Corvigno, S.; Zhong, Y.; Lyons, Y.; Dood, R.; Hu, W.; Qi, L.; Liu, J.; et al. Tumor core biopsies adequately represent immune microenvironment of high-grade serous carcinoma. Sci. Rep. 2019, 9, 17589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hasby, E.A. Weapons ovarian epithelial tumors may use in immune escape: An immunohistochemical correlational study. Pathol. Oncol. Res. 2012, 18, 509–518. [Google Scholar] [CrossRef] [PubMed]
  35. Parkin, J.; Cohen, B. An overview of the immune system. Lancet 2001, 357, 1777–1789. [Google Scholar] [CrossRef]
  36. Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
  37. Tsou, P.; Katayama, H.; Ostrin, E.J.; Hanash, S.M. The Emerging Role of B Cells in Tumor Immunity. Cancer Res. 2016, 76, 5597–5601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Arman Karakaya, Y.; Atıgan, A.; Güler, Ö.; Demiray, A.G.; Bir, F. The relation of CD3, CD4, CD8 and PD-1 expression with tumor type and prognosis in epithelial ovarian cancers. Ginekol. Pol. 2021, 92, 344–351. [Google Scholar] [CrossRef]
  39. Aust, S.; Bachmayr-Heyda, A.; Pils, D.; Zhao, L.; Tong, W.; Berger, A.; Fogel, M.; Thalhammer, T.; Sehouli, J.; Horvat, R.; et al. Determination of tumor-infiltrating CD8+ lymphocytes in human ovarian cancer. Int. J. Gynecol. Pathol. 2013, 32, 269–276. [Google Scholar] [CrossRef]
  40. Bilska, M.; Pawłowska, A.; Zakrzewska, E.; Chudzik, A.; Suszczyk, D.; Gogacz, M.; Wertel, I. Th17 Cells and IL-17 as Novel Immune Targets in Ovarian Cancer Therapy. J. Oncol. 2020, 2020, 15. [Google Scholar] [CrossRef] [Green Version]
  41. Zhu, Q.; Wu, X.; Wang, X. Differential distribution of tumor-associated macrophages and Treg/Th17 cells in the progression of malignant and benign epithelial ovarian tumors. Oncol. Lett. 2017, 13, 159–166. [Google Scholar] [CrossRef]
  42. Webb, J.R.; Wick, D.A.; Nielsen, J.S.; Tran, E.; Milne, K.; McMurtrie, E.; Nelson, B.H. Profound elevation of CD8+ T cells expressing the intraepithelial lymphocyte marker CD103 (αE/β7 Integrin) in high-grade serous ovarian cancer. Gynecol. Oncol. 2010, 118, 228–236. [Google Scholar] [CrossRef] [PubMed]
  43. Oberg, H.H.; Janitschke, L.; Sulaj, V.; Weimer, J.; Gonnermann, D.; Hedemann, N.; Arnold, N.; Kabelitz, D.; Peipp, M.; Bauerschlag, D.; et al. Bispecific antibodies enhance tumor-infiltrating T cell cytotoxicity against autologous HER-2-expressing high-grade ovarian tumors. J. Leukoc. Biol. 2020, 107, 1081–1095. [Google Scholar] [CrossRef] [PubMed]
  44. Robins, H.S.; Ericson, N.G.; Guenthoer, J.; O’Briant, K.C.; Tewari, M.; Drescher, C.W.; Bielas, J.H. Digital genomic quantification of tumor-infiltrating lymphocytes. Sci. Transl. Med. 2013, 5, 214ra169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mhawech-Fauceglia, P.; Wang, D.; Ali, L.; Lele, S.; Huba, M.A.; Liu, S.; Odunsi, K. Intraepithelial T cells and tumor-associated macrophages in ovarian cancer patients. Cancer Immun. 2013, 13, 1. [Google Scholar] [PubMed]
  46. Westergaard, M.C.W.; Andersen, R.; Chong, C.; Kjeldsen, J.W.; Pedersen, M.; Friese, C.; Hasselager, T.; Lajer, H.; Coukos, G.; Bassani-Sternberg, M.; et al. Tumour-reactive T cell subsets in the microenvironment of ovarian cancer. Br. J. Cancer 2019, 120, 424–434. [Google Scholar] [CrossRef] [Green Version]
  47. Landskron, J.; Helland, Ø.; Torgersen, K.M.; Aandahl, E.M.; Gjertsen, B.T.; Bjørge, L.; Taskén, K. Activated regulatory and memory T-cells accumulate in malignant ascites from ovarian carcinoma patients. Cancer Immunol. Immunother. 2015, 64, 337–347. [Google Scholar] [CrossRef]
  48. Kovacsovics-Bankowski, M.; Chisholm, L.; Vercellini, J.; Tucker, C.G.; Montler, R.; Haley, D.; Newell, P.; Ma, J.; Tseng, P.; Wolf, R.; et al. Detailed characterization of tumor infiltrating lymphocytes in two distinct human solid malignancies show phenotypic similarities. J. ImmunoTherapy Cancer 2014, 2, 38. [Google Scholar] [CrossRef]
  49. Barnett, J.C.; Bean, S.M.; Whitaker, R.S.; Kondoh, E.; Baba, T.; Fujii, S.; Marks, J.R.; Dressman, H.K.; Murphy, S.K.; Berchuck, A. Ovarian cancer tumor infiltrating T-regulatory (Treg) cells are associated with a metastatic phenotype. Gynecol. Oncol. 2010, 116, 556–562. [Google Scholar] [CrossRef]
  50. Kroeger, D.R.; Milne, K.; Nelson, B.H. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin. Cancer Res. 2016, 22, 3005–3015. [Google Scholar] [CrossRef] [Green Version]
  51. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef]
  52. Yoshihara, K.; Shahmoradgoli, M.; Martínez, E.; Vegesna, R.; Kim, H.; Torres-Garcia, W.; Treviño, V.; Shen, H.; Laird, P.W.; Levine, D.A.; et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 2013, 4, 2612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Newman, A.M.; Liu, C.L.; Green, M.R.; Gentles, A.J.; Feng, W.; Xu, Y.; Hoang, C.D.; Diehn, M.; Alizadeh, A.A. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 2015, 12, 453–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zhou, X.; Zhao, Y.; Ling, Z.J.; Yang, B. Profiles of immune infiltration in ovarian cancer and their clinical significance: A gene expression-based study. Eur. J. Gynaecol. Oncol. 2021, 42, 346–352. [Google Scholar] [CrossRef]
  55. Gao, Y.; Chen, L.; Cai, G.; Xiong, X.; Wu, Y.; Ma, D.; Li, S.C.; Gao, Q. Heterogeneity of immune microenvironment in ovarian cancer and its clinical significance: A retrospective study. OncoImmunology 2020, 9, 1760067. [Google Scholar] [CrossRef] [PubMed]
  56. Yang, L.; Wang, S.; Zhang, Q.; Pan, Y.; Lv, Y.; Chen, X.; Zuo, Y.; Hao, D. Clinical significance of the immune microenvironment in ovarian cancer patients. Mol. Omics 2018, 14, 341–351. [Google Scholar] [CrossRef] [PubMed]
  57. Murakami, R.; Matsumura, N.; Mandai, M.; Yoshihara, K.; Tanabe, H.; Nakai, H.; Yamanoi, K.; Abiko, K.; Yoshioka, Y.; Hamanishi, J.; et al. Establishment of a Novel Histopathological Classification of High-Grade Serous Ovarian Carcinoma Correlated with Prognostically Distinct Gene Expression Subtypes. Am. J. Pathol. 2016, 186, 1103–1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lakis, S.; Kotoula, V.; Koliou, G.A.; Efstratiou, I.; Chrisafi, S.; Papanikolaou, A.; Zebekakis, P.; Fountzilas, G. Multisite Tumor Sampling Reveals Extensive Heterogeneity of Tumor and Host Immune Response in Ovarian Cancer. Cancer Genom. Proteom. 2020, 17, 529–541. [Google Scholar] [CrossRef]
  59. Kreuzinger, C.; Geroldinger, A.; Smeets, D.; Braicu, E.I.; Sehouli, J.; Koller, J.; Wolf, A.; Darb-Esfahani, S.; Joehrens, K.; Vergote, I.; et al. A complex network of tumor microenvironment in human high-grade serous ovarian cancer. Clin. Cancer Res. 2017, 23, 7621–7632. [Google Scholar] [CrossRef] [Green Version]
  60. Ojalvo, L.S.; Thompson, E.D.; Wang, T.L.; Meeker, A.K.; Shih, I.M.; Fader, A.N.; Cimino-Mathews, A.; Emens, L.A. Tumor-associated macrophages and the tumor immune microenvironment of primary and recurrent epithelial ovarian cancer. Hum. Pathol. 2018, 74, 135–147. [Google Scholar] [CrossRef]
  61. Olalekan, S.; Xie, B.; Back, R.; Eckart, H.; Basu, A. Characterizing the tumor microenvironment of metastatic ovarian cancer by single-cell transcriptomics. Cell Rep. 2021, 35, 109165. [Google Scholar] [CrossRef]
  62. Zhu, Y.; Ferri-Borgogno, S.; Sheng, J.; Yeung, T.L.; Burks, J.K.; Cappello, P.; Jazaeri, A.A.; Kim, J.H.; Han, G.H.; Birrer, M.J.; et al. Sio: A spatioimageomics pipeline to identify prognostic biomarkers associated with the ovarian tumor microenvironment. Cancers 2021, 13, 1777. [Google Scholar] [CrossRef] [PubMed]
  63. Hagemann, A.R.; Hagemann, I.S.; Cadungog, M.; Hwang, W.T.; Patel, P.; Lal, P.; Hammond, R.; Gimotty, P.A.; Chu, C.S.; Rubin, S.C.; et al. Tissue-based immune monitoring II: Multiple tumor sites reveal immunologic homogeneity in serous ovarian carcinoma. Cancer Biol. Ther. 2011, 12, 367–377. [Google Scholar] [CrossRef] [Green Version]
  64. Ojalvo, L.S.; Thompson, E.D.; Wang, T.L.; Meeker, A.K.; Shih, I.M.; Fader, A.N.; Cimino-Mathews, A.; Emens, L.A. Profiling the immune tumor microenvironment in primary and recurrent epithelial ovarian cancer. Cancer Res. 2017, 77, 3991. [Google Scholar] [CrossRef]
  65. Nakamura, M.; Bax, H.J.; Scotto, D.; Souri, E.A.; Sollie, S.; Harris, R.J.; Hammar, N.; Walldius, G.; Winship, A.; Ghosh, S.; et al. Immune mediator expression signatures are associated with improved outcome in ovarian carcinoma. OncoImmunology 2019, 8, e1593811. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. James, F.R.; Jiminez-Linan, M.; Alsop, J.; Mack, M.; Song, H.; Brenton, J.D.; Pharoah, P.D.P.; Ali, H.R. Association between tumour infiltrating lymphocytes, histotype and clinical outcome in epithelial ovarian cancer. BMC Cancer 2017, 17, 657. [Google Scholar] [CrossRef]
  68. Hwang, C.; Lee, S.J.; Lee, J.H.; Kim, K.H.; Suh, D.S.; Kwon, B.S.; Choi, K.U. Stromal tumor-infiltrating lymphocytes evaluated on H&E-stained slides are an independent prognostic factor in epithelial ovarian cancer and ovarian serous carcinoma. Oncol. Lett. 2019, 17, 4557–4565. [Google Scholar] [CrossRef] [Green Version]
  69. Bansal, A.; Srinivasan, R.; Rohilla, M.; Rai, B.; Rajwanshi, A.; Suri, V.; Chandra Saha, S. Immunotyping in tubo-ovarian high-grade serous carcinoma by PD-L1 and CD8+ T-lymphocytes predicts disease-free survival. APMIS 2021, 129, 254–264. [Google Scholar] [CrossRef]
  70. Sato, E.; Olson, S.H.; Ahn, J.; Bundy, B.; Nishikawa, H.; Qian, F.; Jungbluth, A.A.; Frosina, D.; Gnjatic, S.; Ambrosone, C.; et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 18538–18543. [Google Scholar] [CrossRef] [Green Version]
  71. Hamanishi, J.; Mandai, M.; Iwasaki, M.; Okazaki, T.; Tanaka, Y.; Yamaguchi, K.; Higuchi, T.; Yagi, H.; Takakura, K.; Minato, N.; et al. Programmed Cell Death 1 Ligand 1 and Tumor-Infiltrating CD8⁺ T Lymphocytes Are Prognostic Factors of Human Ovarian Cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 3360–3365. [Google Scholar] [CrossRef]
  72. Kryczek, I.; Banerjee, M.; Cheng, P.; Vatan, L.; Szeliga, W.; Wei, S.; Huang, E.; Finlayson, E.; Simeone, D.; Welling, T.H.; et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood 2009, 114, 1141–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
  74. Leffers, N.; Gooden, M.J.M.; de Jong, R.A.; Hoogeboom, B.-N.; ten Hoor, K.A.; Hollema, H.; Boezen, H.M.; van der Zee, A.G.J.; Daemen, T.; Nijman, H.W. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol. Immunother. 2009, 58, 449–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Milne, K.; Köbel, M.; Kalloger, S.E.; Barnes, R.O.; Gao, D.; Gilks, C.B.; Watson, P.H.; Nelson, B.H. Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS ONE 2009, 4, e6412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tsuji, T.; Eng, K.H.; Matsuzaki, J.; Battaglia, S.; Brian Szender, J.; Miliotto, A.; Gnjatic, S.; Bshara, W.; Morrison, C.D.; Lele, S.; et al. Clonality and antigen-specific responses shape the prognostic effects of tumor-infiltrating T cells in ovarian cancer. Oncotarget 2020, 11, 2669–2683. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, G.; Xu, Q.; Zhang, X.; Yang, M.; Wang, Y.; He, M.; Lu, J.; Liu, H. Spatial cytotoxic and memory T cells in tumor predict superior survival outcomes in patients with high-grade serous ovarian cancer. Cancer Med. 2021, 10, 3905–3918. [Google Scholar] [CrossRef]
  78. Han, G.H.; Hwang, I.; Cho, H.; Ylaya, K.; Choi, J.A.; Kwon, H.; Chung, J.Y.; Hewitt, S.M.; Kim, J.H. Clinical Significance of Tumor Infiltrating Lymphocytes in Association with Hormone Receptor Expression Patterns in Epithelial Ovarian Cancer. Int. J. Mol. Sci. 2021, 22, 5714. [Google Scholar] [CrossRef]
  79. De La Motte Rouge, T.; Corné, J.; Cauchois, A.; Le Boulch, M.; Poupon, C.; Henno, S.; Rioux-Leclercq, N.; Le Pabic, E.; Laviolle, B.; Catros, V.; et al. Serum CD95L level correlates with tumor immune infiltration and is a positive prognostic marker for advanced high-grade serous ovarian cancer. Mol. Cancer Res. 2019, 17, 2537–2548. [Google Scholar] [CrossRef] [Green Version]
  80. Preston, C.; Maurer, M.; Oberg, A.; Hartmann, L.; Kalli, K.; Goode, E.; Knutson, K. CD4+CD25+FOXP3+ regulatory T cells and association with survival in epithelial ovarian cancer. J. Immunol. 2012, 188, 1. [Google Scholar]
  81. Santoiemma, P.P.; Reyes, C.; Wang, L.P.; McLane, M.W.; Feldman, M.D.; Tanyi, J.L.; Powell, D.J. Systematic evaluation of multiple immune markers reveals prognostic factors in ovarian cancer. Gynecol. Oncol. 2016, 143, 120–127. [Google Scholar] [CrossRef]
  82. Yildirim, N.; Akman, L.; Acar, K.; Demir, S.; Ozkan, S.; Alan, N.; Zekioglu, O.; Terek, M.C.; Ozdemir, N.; Ozsaran, A. Do tumor-infiltrating lymphocytes really indicate favorable prognosis in epithelial ovarian cancer? EJOG 2017, 215, 55–61. [Google Scholar] [CrossRef] [PubMed]
  83. Hinchcliff, E.M.; Paquette, C.; Roszik, J.; Kelting, S.; Stoler, M.H.; Mok, S.C.; Yeung, T.; Zhang, Q.; Yates, M.; Peng, W.; et al. Lymphocyte-specific protein tyrosine kinase expression predicts survival in ovarian high-grade serous carcinoma. Gynecol. Oncol. 2019, 154, 64–65. [Google Scholar] [CrossRef]
  84. Martin de la Fuente, L.; Westbom-Fremer, S.; Arildsen, N.S.; Hartman, L.; Malander, S.; Kannisto, P.; Måsbäck, A.; Hedenfalk, I. PD-1/PD-L1 expression and tumor-infiltrating lymphocytes are prognostically favorable in advanced high-grade serous ovarian carcinoma. Virchows Arch. 2020, 477, 83–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Stanske, M.; Wienert, S.; Castillo-Tong, D.C.; Kreuzinger, C.; Vergote, I.; Lambrechts, S.; Gabra, H.; Gourley, C.; Ganapathi, R.N.; Kolaschinski, I.; et al. Dynamics of the Intratumoral Immune Response during Progression of High-Grade Serous Ovarian Cancer. Neoplasia 2018, 20, 280–288. [Google Scholar] [CrossRef]
  86. Wang, S.; Wang, C.; Liu, O.; Hu, Y.; Li, X.; Lin, B. Prognostic value of immune-related cells and genes in the tumor microenvironment of ovarian cancer, especially CST4. Life Sci. 2021, 277, 119461. [Google Scholar] [CrossRef]
  87. Pinto, M.P.; Balmaceda, C.; Bravo, M.L.; Kato, S.; Villarroel, A.; Owen, G.I.; Roa, J.C.; Cuello, M.A.; Ibañez, C. Patient inflammatory status and CD4+/CD8+ intraepithelial tumor lymphocyte infiltration are predictors of outcomes in high-grade serous ovarian cancer. Gynecol. Oncol. 2018, 151, 10–17. [Google Scholar] [CrossRef] [Green Version]
  88. Hinchcliff, E.; Paquette, C.; Roszik, J.; Kelting, S.; Stoler, M.H.; Mok, S.C.; Yeung, T.L.; Zhang, Q.; Yates, M.; Peng, W.; et al. Lymphocyte-specific kinase expression is a prognostic indicator in ovarian cancer and correlates with a prominent B cell transcriptional signature. Cancer Immunol. Immunother. 2019, 68, 1515–1526. [Google Scholar] [CrossRef]
  89. Nielsen, J.S.; Sahota, R.A.; Milne, K.; Kost, S.E.; Nesslinger, N.J.; Watson, P.H.; Nelson, B.H. CD20+ tumor-infiltrating lymphocytes have an atypical CD27—Memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin. Cancer Res. 2012, 18, 3281–3292. [Google Scholar] [CrossRef] [Green Version]
  90. Hermans, C.; Anz, D.; Engel, J.; Kirchner, T.; Endres, S.; Mayr, D. Analysis of FoxP3+ T-regulatory cells and CD8+T-Cells in ovarian carcinoma: Location and tumor infiltration patterns are key prognostic markers. PLoS ONE 2014, 9, e111757. [Google Scholar] [CrossRef] [Green Version]
  91. De Leeuw, R.J.; Kroeger, D.R.; Kost, S.E.; Chang, P.P.; Webb, J.R.; Nelson, B.H. CD25 identifies a subset of CD4 FoxP3 TIL that are exhausted yet prognostically favorable in Human Ovarian cancer. Cancer Immunol. Res. 2015, 3, 245–253. [Google Scholar] [CrossRef] [Green Version]
  92. Farrag, M.S.; Abdelwahab, K.; Farrag, N.S.; Elrefaie, W.E.; Emarah, Z. Programmed death ligand-1 and CD8 tumor-infiltrating lymphocytes (TILs) as prognostic predictors in ovarian high-grade serous carcinoma (HGSC). J. Egypt. Natl. Cancer Inst. 2021, 33, 16. [Google Scholar] [CrossRef] [PubMed]
  93. Bachmayr-Heyda, A.; Aust, S.; Heinze, G.; Polterauer, S.; Grimm, C.; Braicu, E.I.; Sehouli, J.; Lambrechts, S.; Vergote, I.; Mahner, S.; et al. Prognostic impact of tumor infiltrating CD8+ T cells in association with cell proliferation in ovarian cancer patients—A study of the OVCAD consortium. BMC Cancer 2013, 13, 422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Preston, C.C.; Maurer, M.J.; Oberg, A.L.; Visscher, D.W.; Kalli, K.R.; Hartmann, L.C.; Goode, E.L.; Knutson, K.L. The ratios of CD8+ T cells to CD4+CD25+ FOXP3+ and FOXP3- T cells correlate with poor clinical outcome in human serous ovarian cancer. PLoS ONE 2013, 8, e80063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Laumont, C.M.; Wouters, M.C.A.; Smazynski, J.; Gierc, N.S.; Chavez, E.A.; Chong, L.C.; Thornton, S.; Milne, K.; Webb, J.R.; Steidl, C.; et al. Single-cell profiles and prognostic impact of tumor-infiltrating lymphocytes coexpressing CD39, CD103, and PD-1 in ovarian cancer. Clin. Cancer Res. 2021, 27, 4089–4100. [Google Scholar] [CrossRef] [PubMed]
  96. Webb, J.R.; Milne, K.; Watson, P.; DeLeeuw, R.J.; Nelson, B.H. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker cd103 are associated with increased survival in high-grade serous ovarian cancer. Clin. Cancer Res. 2014, 20, 434–444. [Google Scholar] [CrossRef] [Green Version]
  97. Paijens, S.T.; Vledder, A.; Loiero, D.; Duiker, E.W.; Bart, J.; Hendriks, A.M.; Jalving, M.; Workel, H.H.; Hollema, H.; Werner, N.; et al. Prognostic image-based quantification of CD8CD103 T cell subsets in high-grade serous ovarian cancer patients. Oncoimmunology 2021, 10, 1935104. [Google Scholar] [CrossRef]
  98. Khalique, S.; Nash, S.; Mansfield, D.; Wampfler, J.; Attygale, A.; Vroobel, K.; Kemp, H.; Buus, R.; Cottom, H.; Roxanis, I.; et al. Quantitative Assessment and Prognostic Associations of the Immune Landscape in Ovarian Clear Cell Carcinoma. Cancers 2021, 13, 3854. [Google Scholar] [CrossRef]
  99. Iglesia, M.D.; Vincent, B.G.; Parker, J.S.; Hoadley, K.A.; Carey, L.A.; Perou, C.M.; Serody, J.S. Prognostic B-cell signatures using mRNA-seq in patients with subtype-specific breast and ovarian cancer. Clin. Cancer Res. 2014, 20, 3818–3829. [Google Scholar] [CrossRef] [Green Version]
  100. Lundgren, S.; Berntsson, J.; Nodin, B.; Micke, P.; Jirström, K. Prognostic impact of tumour-associated B cells and plasma cells in epithelial ovarian cancer. J. Ovarian Res. 2016, 9, 21. [Google Scholar] [CrossRef] [Green Version]
  101. Webb, J.R.; Milne, K.; Nelson, B.H. Location, location, location: CD103 demarcates intraepithelial, prognostically favorable CD8+ tumor-infiltrating lymphocytes in ovarian cancer. Oncoimmunology 2014, 3, e27668. [Google Scholar] [CrossRef] [Green Version]
  102. Wu, Y.; Gao, Y.; Chen, L.; Jin, X.; Chen, P.; Mo, Q. Prognostic implications of tumour-infiltrating lymphocytes for recurrence in epithelial ovarian cancer. Clin. Exp. Immunol. 2021, 206, 36–46. [Google Scholar] [CrossRef] [PubMed]
  103. Li, N.; Li, B.; Zhan, X. Comprehensive Analysis of Tumor Microenvironment Identified Prognostic Immune-Related Gene Signature in Ovarian Cancer. Front. Genet. 2021, 12, 616073. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, H.; Molberg, K.; Strickland, A.L.; Castrillon, D.H.; Carrick, K.; Jiang, Q.; Niu, S.; Rivera-Colon, G.; Gwin, K.; Hinson, S.; et al. PD-L1 Expression and CD8+ Tumor-infiltrating Lymphocytes in Different Types of Tubo-ovarian Carcinoma and Their Prognostic Value in High-grade Serous Carcinoma. Am. J. Surg. Pathol. 2020, 44, 1050–1060. [Google Scholar] [CrossRef] [PubMed]
  105. Xiao, X.; Dong, D.; He, W.; Song, L.; Wang, Q.; Yue, J.; Xie, L. Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer. Gynecol. Oncol. 2018, 149, 146–154. [Google Scholar] [CrossRef]
  106. Testa, U.; Petrucci, E.; Pasquini, L.; Castelli, G.; Pelosi, E. Ovarian Cancers: Genetic Abnormalities, Tumor Heterogeneity and Progression, Clonal Evolution and Cancer Stem Cells. Medicines 2018, 5, 16. [Google Scholar] [CrossRef] [Green Version]
  107. Zhao, Y.; Yu, Z.; Ma, R.; Zhang, Y.; Zhao, L.; Yan, Y.; Lv, X.; Zhang, L.; Su, P.; Bi, J.; et al. lncRNA-Xist/miR-101-3p/KLF6/C/EBPα axis promotes TAM polarization to regulate cancer cell proliferation and migration. Mol. Ther.—Nucleic Acids 2021, 23, 536–551. [Google Scholar] [CrossRef]
  108. Matsushita, H.; Hasegawa, K.; Oda, K.; Yamamoto, S.; Asada, K.; Karasaki, T.; Yabuno, A.; Nishijima, A.; Nejo, T.; Kobayashi, Y.; et al. Neoantigen load and HLA-class i expression identify a subgroup of tumors with a T-cell-inflamed phenotype and favorable prognosis in homologous recombination-proficient high-grade serous ovarian carcinoma. J. Immunother. Cancer 2020, 8, e000375. [Google Scholar] [CrossRef]
  109. Strickland, K.C.; Howitt, B.E.; Shukla, S.A.; Rodig, S.; Ritterhouse, L.L.; Liu, J.F.; Garber, J.E.; Chowdhury, D.; Wu, C.J.; D’Andrea, A.D.; et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016, 7, 13587–13598. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, J.; Zhang, T.; Yang, L.; Yang, G. Comprehensive genomic analysis of microenvironment phenotypes in ovarian cancer. PeerJ 2020, 8, e10255. [Google Scholar] [CrossRef]
  111. Morse, C.B.; Toukatly, M.N.; Kilgore, M.R.; Agnew, K.J.; Bernards, S.S.; Norquist, B.M.; Pennington, K.P.; Garcia, R.L.; Liao, J.B.; Swisher, E.M. Tumor infiltrating lymphocytes and homologous recombination deficiency are independently associated with improved survival in ovarian carcinoma. Gynecol. Oncol. 2019, 153, 217–222. [Google Scholar] [CrossRef]
  112. Wei, Y.; Ou, T.; Lu, Y.; Wu, G.; Long, Y.; Pan, X.; Yao, D. Classification of ovarian cancer associated with BRCA1 mutations, immune checkpoints, and tumor microenvironment based on immunogenomic profiling. PeerJ 2020, 8, e10414. [Google Scholar] [CrossRef] [PubMed]
  113. Rasmussen, M.; Lim, K.; Rambech, E.; Andersen, M.H.; Svane, I.M.; Andersen, O.; Jensen, L.H.; Nilbert, M.; Therkildsen, C. Lynch syndrome-associated epithelial ovarian cancer and its immunological profile. Gynecol. Oncol. 2021, 162, 686–693. [Google Scholar] [CrossRef] [PubMed]
  114. Whitehair, R.; Peres, L.C.; Mills, A.M. Expression of the Immune Checkpoints LAG-3 and PD-L1 in High-grade Serous Ovarian Carcinoma: Relationship to Tumor-associated Lymphocytes and Germline BRCA Status. Int. J. Gynecol. Pathol. 2020, 39, 558–566. [Google Scholar] [CrossRef]
  115. Heindl, A.; Khan, A.M.; Rodrigues, D.N.; Eason, K.; Sadanandam, A.; Orbegoso, C.; Punta, M.; Sottoriva, A.; Lise, S.; Banerjee, S.; et al. Microenvironmental niche divergence shapes BRCA1-dysregulated ovarian cancer morphological plasticity. Nat. Commun. 2018, 9, 3917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Coukos, G.; Tanyi, J.; Kandalaft, L.E. Opportunities in immunotherapy of ovarian cancer. Ann. Oncol. 2016, 27, i11–i15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [Green Version]
  118. Chang, E.; Pelosof, L.; Lemery, S.; Gong, Y.; Goldberg, K.B.; Farrell, A.T.; Keegan, P.; Veeraraghavan, J.; Wei, G.; Blumenthal, G.M.; et al. Systematic Review of PD-1/PD-L1 Inhibitors in Oncology: From Personalized Medicine to Public Health. Oncologist 2021, 26, e1786–e1799. [Google Scholar] [CrossRef]
  119. Davis, A.A.; Patel, V.G. The role of PD-L1 expression as a predictive biomarker: An analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef]
  120. Webb, J.R.; Milne, K.; Nelson, B.H. PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer. Cancer Immunol. Res. 2015, 3, 926–935. [Google Scholar] [CrossRef] [Green Version]
  121. Webb, J.R.; Milne, K.; Kroeger, D.R.; Nelson, B.H. PD-L1 expression is associated with tumor-infiltrating T cells and favorable prognosis in high-grade serous ovarian cancer. Gynecol. Oncol. 2016, 141, 293–302. [Google Scholar] [CrossRef] [Green Version]
  122. Darb-Esfahani, S.; Kunze, C.A.; Kulbe, H.; Sehouli, J.; Wienert, S.; Lindner, J.; Budczies, J.; Bockmayr, M.; Dietel, M.; Denkert, C.; et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumorinfiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget 2016, 7, 1486–1499. [Google Scholar] [CrossRef] [PubMed]
  123. Fucikova, J.; Rakova, J.; Hensler, M.; Kasikova, L.; Belicova, L.; Hladikova, K.; Truxova, I.; Skapa, P.; Laco, J.; Pecen, L.; et al. TIM-3 dictates functional orientation of the immune infiltrate in ovarian cancer. Clin. Cancer Res. 2019, 25, 4820–4831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wang, Q.; Lou, W.; Di, W.; Wu, X. Prognostic value of tumor PD-L1 expression combined with CD8(+) tumor infiltrating lymphocytes in high grade serous ovarian cancer. Int. Immunopharmacol. 2017, 52, 7–14. [Google Scholar] [CrossRef] [PubMed]
  125. Kim, K.H.; Choi, K.U.; Kim, A.; Lee, S.J.; Lee, J.H.; Suh, D.S.; Kwon, B.S.; Hwang, C. PD-L1 expression on stromal tumor-infiltrating lymphocytes is a favorable prognostic factor in ovarian serous carcinoma. J. Ovarian Res. 2019, 12, 56. [Google Scholar] [CrossRef] [PubMed]
  126. Sawada, M.; Goto, K.; Morimoto-Okazawa, A.; Haruna, M.; Yamamoto, K.; Yamamoto, Y.; Nakagawa, S.; Hiramatsu, K.; Matsuzaki, S.; Kobayashi, E.; et al. PD-1+Tim3+tumor-infiltrating CD8 T cells sustain the potential for IFN-γproduction, but lose cytotoxic activity in ovarian cancer. Int. Immunol. 2020, 32, 397–405. [Google Scholar] [CrossRef]
  127. Bekos, C.; Pils, D.; Dekan, S.; Hofstetter, G.; Horak, P.; Reinthaller, A.; Polterauer, S.; Schwameis, R.; Aust, S. PD-1 and PD-L1 expression on TILs in peritoneal metastases compared to ovarian tumor tissues and its associations with clinical outcome. Sci. Rep. 2021, 11, 6400. [Google Scholar] [CrossRef]
  128. Wolf, Y.; Anderson, A.C.; Kuchroo, V.K. TIM3 comes of age as an inhibitory receptor. Nat. Rev. Immunol. 2020, 20, 173–185. [Google Scholar] [CrossRef]
  129. Sakellariou-Thompson, D.; Forget, M.A.; Hinchcliff, E.; Celestino, J.; Hwu, P.; Jazaeri, A.A.; Haymaker, C.; Bernatchez, C. Potential clinical application of tumor-infiltrating lymphocyte therapy for ovarian epithelial cancer prior or post-resistance to chemotherapy. Cancer Immunol. Immunother. 2019, 68, 1747–1757. [Google Scholar] [CrossRef]
  130. Böhm, S.; Montfort, A.; Pearce, O.M.T.; Topping, J.; Chakravarty, P.; Everitt, G.L.A.; Clear, A.; McDermott, J.R.; Ennis, D.; Dowe, T.; et al. Neoadjuvant chemotherapy modulates the immune microenvironment in metastases of tubo-ovarian high-grade serous carcinoma. Clin. Cancer Res. 2016, 22, 3025–3036. [Google Scholar] [CrossRef] [Green Version]
  131. van Baal, J.O.A.M.; Lok, C.A.R.; Jordanova, E.S.; Horlings, H.; van Driel, W.J.; Amant, F.C.; Van de Vijver, K.K. The effect of the peritoneal tumor microenvironment on invasion of peritoneal metastases of high-grade serous ovarian cancer and the impact of NEOADJUVANT chemotherapy. Virchows Arch. 2020, 477, 535–544. [Google Scholar] [CrossRef]
  132. Pölcher, M.; Braun, M.; Friedrichs, N.; Rudlowski, C.; Bercht, E.; Fimmers, R.; Sauerwald, A.; Keyver-Paik, M.D.; Kübler, K.; Büttner, R.; et al. Foxp3+ cell infiltration and granzyme B+/Foxp3 + cell ratio are associated with outcome in neoadjuvant chemotherapy-treated ovarian carcinoma. Cancer Immunol. Immunother. 2010, 59, 909–919. [Google Scholar] [CrossRef] [PubMed]
  133. Böhm, S.; Le, N.; Lockley, M.; Brockbank, E.; Faruqi, A.; Said, I.; Jeyarajah, A.; Wuntakal, R.; Gilks, B.; Singh, N. Histopathologic response to neoadjuvant chemotherapy as a prognostic biomarker in tubo-ovarian high-grade serous carcinoma: Updated Chemotherapy Response Score (CRS) results. Int. J. Gynecol. Cancer 2019, 29, 353–356. [Google Scholar] [CrossRef] [PubMed]
  134. Mesnage, S.J.L.; Auguste, A.; Genestie, C.; Dunant, A.; Pain, E.; Drusch, F.; Gouy, S.; Morice, P.; Bentivegna, E.; Lhomme, C.; et al. Neoadjuvant chemotherapy (NACT) increases immune infiltration and programmed death-ligand 1 (PD-L1) expression in epithelial ovarian cancer (EOC). Ann. Oncol. 2017, 28, 651–657. [Google Scholar] [CrossRef] [PubMed]
  135. Lake, R.A.; Robinson, B.W. Immunotherapy and chemotherapy—A practical partnership. Nat. Rev. Cancer 2005, 5, 397–405. [Google Scholar] [CrossRef] [PubMed]
  136. Ghiringhelli, F.; Larmonier, N.; Schmitt, E.; Parcellier, A.; Cathelin, D.; Garrido, C.; Chauffert, B.; Solary, E.; Bonnotte, B.; Martin, F. CD4⁺CD25⁺ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 2004, 34, 336–344. [Google Scholar] [CrossRef]
  137. Leary, A.; Genestie, C.; Blanc-Durand, F.; Gouy, S.; Dunant, A.; Maulard, A.; Drusch, F.; Cheaib, B.; Michels, J.; Bentivegna, E.; et al. Neoadjuvant chemotherapy alters the balance of effector to suppressor immune cells in advanced ovarian cancer. Cancer Immunol. Immunother. 2021, 70, 519–531. [Google Scholar] [CrossRef]
  138. Bobisse, S.; Genolet, R.; Roberti, A.; Tanyi, J.L.; Racle, J.; Stevenson, B.J.; Iseli, C.; Michel, A.; Le Bitoux, M.A.; Guillaume, P.; et al. Sensitive and frequent identification of high avidity neo-epitope specific CD8+ T cells in immunotherapy-naive ovarian cancer. Nat. Commun. 2018, 9, 1092. [Google Scholar] [CrossRef] [Green Version]
  139. Kandalaft, L.E.; Odunsi, K.; Coukos, G. Immune Therapy Opportunities in Ovarian Cancer. Am. Soc. Clin. Oncol. Educ. Book 2020, 40, 1–13. [Google Scholar] [CrossRef]
  140. Sarivalasis, A.; Morotti, M.; Mulvey, A.; Imbimbo, M.; Coukos, G. Cell therapies in ovarian cancer. Ther. Adv. Med. Oncol. 2021, 13, 17588359211008399. [Google Scholar] [CrossRef]
  141. Aust, S.; Schwameis, R.; Gagic, T.; Müllauer, L.; Langthaler, E.; Prager, G.; Grech, C.; Reinthaller, A.; Krainer, M.; Pils, D.; et al. Precision Medicine Tumor Boards: Clinical Applicability of Personalized Treatment Concepts in Ovarian Cancer. Cancers 2020, 12, 548. [Google Scholar] [CrossRef] [Green Version]
  142. Herrera, F.G.; Irving, M.; Kandalaft, L.E.; Coukos, G. Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol. 2019, 20, e417–e433. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of the selection process for the studies included in this review. Review according to Moher [21], EOC: epithelial ovarian cancer.
Figure 1. Schematic diagram of the selection process for the studies included in this review. Review according to Moher [21], EOC: epithelial ovarian cancer.
Cancers 14 05332 g001
Figure 2. Immunologic network in EOC: (A) simplified TIL view and location; (B) Main immune checkpoints studied in EOC. Simplified diagram of the main TILs described in the articles studied in this review. T cells infiltrating the stroma or tumor epithelium are identified via CD3, and/or CD4 and CD8. The subtypes of T cells, including TH1, TH2, TH17, TFH, and TREG, are illustrated. The main immune checkpoints described in this review are represented. iTILs: intra-tumoral, sTILs: stromal, B-TILs: B tumor-infiltrating lymphocytes, TCR: T cell receptor, PD-1: programmed-death 1, PD-L1: PD-1 ligand 1, PD-L2: PD-1 ligand 2, CTLA4: cytotoxic T-lymphocyte-associated protein 4, Tim-3: T cell immunoglobulin and mucin domain-containing protein 3, and LAG-3: lymphocyte activating gene 3.
Figure 2. Immunologic network in EOC: (A) simplified TIL view and location; (B) Main immune checkpoints studied in EOC. Simplified diagram of the main TILs described in the articles studied in this review. T cells infiltrating the stroma or tumor epithelium are identified via CD3, and/or CD4 and CD8. The subtypes of T cells, including TH1, TH2, TH17, TFH, and TREG, are illustrated. The main immune checkpoints described in this review are represented. iTILs: intra-tumoral, sTILs: stromal, B-TILs: B tumor-infiltrating lymphocytes, TCR: T cell receptor, PD-1: programmed-death 1, PD-L1: PD-1 ligand 1, PD-L2: PD-1 ligand 2, CTLA4: cytotoxic T-lymphocyte-associated protein 4, Tim-3: T cell immunoglobulin and mucin domain-containing protein 3, and LAG-3: lymphocyte activating gene 3.
Cancers 14 05332 g002
Figure 3. Synthesis of the effects of TILs on prognosis in EOC. This figure summarizes the conclusion of the articles exploring the effects of TILs on EOC prognosis, either being evaluated in HES or via the study of a surface marker. B-TILs are mostly identified using CD20. H&E: hematoxylin and eosin, and B-TILs: B tumor-infiltrating lymphocytes.
Figure 3. Synthesis of the effects of TILs on prognosis in EOC. This figure summarizes the conclusion of the articles exploring the effects of TILs on EOC prognosis, either being evaluated in HES or via the study of a surface marker. B-TILs are mostly identified using CD20. H&E: hematoxylin and eosin, and B-TILs: B tumor-infiltrating lymphocytes.
Cancers 14 05332 g003
Table 3. TILs and immune checkpoints in the reviewed studies.
Table 3. TILs and immune checkpoints in the reviewed studies.
Study Author (Publication Year)Number of Cases n=PD-1PD-L1Definition of Positive TILsScoring PD-1Scoring PD-L1Method
Webb (2015) [120]489PD-1+ cells: positive factor DFS in HGSOC, and not in other EOCsNot studiedCD3 and CD8 >5 and < 5absolute numbers of PD-1+ and CD103+Not studiedimmunohistochemistry, flow cytometry
Webb (2016) [121]490Not studiedPD-L1 expression: positive factor OS in HGSOC, and no difference in other EOCsCD8 quantitative pathology imaging systemNot studiedPD-L1 scored as positive or negative, using a threshold of ≥1 positive cellsimmunohistochemistry, TCGA
Darb Esfahni (2016) [122]215PD-1 on cancer cells and TILs: positive factor OSPD-L1 on cancer cells and TILs: positive factor DFS and OSi and sCD3 cut-off: > 65/mm2cancer cells PD-1+ > 11/mm2cancer cells PD-L1+ > 20/mm2Tissue microarrays + MRNA expression + MA
Wang (2017) [124]107Not studiedtumor PD-L1 expression: negative factor for OS; TILs-PD-L1+: no difference for OSsTILs: score 1 (≤5/HPF), 2 (6–20/HPF), and 3 (≥20/HPF); iTILs: score 1 (≤5/HPF) and 2 (>5/HPF)Not studiedPD-L1 staining in tumor cells scored: 0, negative; 1, weak expression; 2, moderate expression but weaker than placenta; and 3, equivalent or stronger expression than placenta.immunohistochemistry
Fucikova (2019) [123]80PD-1 high: positive factor OSPD-L1 (positive vs negative): positive factor OSCD8: entire TME: absolute number of positive cells/mm2; CD20: cell surface/tumor section surfacePD-1, CTLA4, LAG-3: stroma and tumor of whole tumorPD-L1 intratumoral and stromal, categorized as 1 (0%), 2 (1–4%), 4 (5–9%), and 4 (>10%); cut-off 5% to survival analysisimmunohistochemistry + flow cytometry + TCGA
Kim (2019) [125]248Not studiedsTILs-PD-L1+: positive factor OSstromal sTILs, and iTILs: graded on a semiquantitative scale of 0 (none), 1+ (mild), 2+ (moderate), and 3+ (marked)Not studiedintensities of PD-L1: intraepithelial (staining in tumor cells)immunohistochemistry
Martin de la Fuente (2020) [84]130high PD-1 expression: better OShigh PD-L1 expression: better OSgrading CD3: 0%, < 1%, 1%, 2–4%, and ≥ 5% (high expression = ≥ 50% cores with ≥ 2% lymphocyte)PD-1 expression ≥ 1% in ≤50% cores considered high expressiongrading PD-L1 and PD-L1: 0%, <1%, 1–4%, ≥5%; iTILs PD-1Tissue microarray construction
and immunohistochemistry
Chen (2020) [104]189Not studiedin HGSOC TPS: better DFS and OS (CPS: no difference) Not studiedTPS and CPS; TPS and CPS ranged from 0 to 100; cutoff score ≥1% for TPS and ≥1 for CPS used to define PD-L1 positivity; for CPS intratumoral and peritumoral, stromal immune cells excludedimmunohistochemistry
Bekos (2021) [127]111PD-1 in TILs in peritoneal metastases: positive factor OSPD-L1 in TILs in peritoneal metastases: negative factor OSCD8+: % (cut-off 44.3%)PD-1 % in TILs (cut-off 40%)PD-L1 % in TILs: ovarian tissue and peritoneal samples (cut-off 15%)immunohistochemistry
Bansal (2021) [69]100Not studiedCPS or TCS PD-L1: no correlation with DFSsTILs and iTILs: quantified in 5 different (400×) HPF on 3 sections/cases, 0: no lymphoid cells, 1: mild, 2: moderate, and 3: numerous numbersNot studiedCPSimmunohistochemistry
This review brings together the articles about PD-1 and PD-L1, two preferential ICI targets in EOC. The cut-off, the way of estimating the markers’ expression, and the method used are identified for each study. TPS: tumor proportion score: % tumor cells with membranous PD-L1 expression; CPS: combined positive score: (tumor cell, lymphocyte, and macrophage PD-L1 staining cells)/(total number of viable tumor cells) × 100; EOC: epithelial ovarian cancer; HGSOC high-grade serous ovarian cancer; MA: multi analyze; HPF: high power fields; DFS: disease free survival; and OS: overall survival.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hudry, D.; Le Guellec, S.; Meignan, S.; Bécourt, S.; Pasquesoone, C.; El Hajj, H.; Martínez-Gómez, C.; Leblanc, É.; Narducci, F.; Ladoire, S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers 2022, 14, 5332. https://doi.org/10.3390/cancers14215332

AMA Style

Hudry D, Le Guellec S, Meignan S, Bécourt S, Pasquesoone C, El Hajj H, Martínez-Gómez C, Leblanc É, Narducci F, Ladoire S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers. 2022; 14(21):5332. https://doi.org/10.3390/cancers14215332

Chicago/Turabian Style

Hudry, Delphine, Solenn Le Guellec, Samuel Meignan, Stéphanie Bécourt, Camille Pasquesoone, Houssein El Hajj, Carlos Martínez-Gómez, Éric Leblanc, Fabrice Narducci, and Sylvain Ladoire. 2022. "Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints" Cancers 14, no. 21: 5332. https://doi.org/10.3390/cancers14215332

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop