The expression of programmed cell death ligand 1 (PD-L1) involves in the clinicopathologic characteristics and prognostic implications of testicular germ cell tumor (TGCT): a systematic review and meta-analysis
Original Article

The expression of programmed cell death ligand 1 (PD-L1) involves in the clinicopathologic characteristics and prognostic implications of testicular germ cell tumor (TGCT): a systematic review and meta-analysis

Peifeng Li1,2#, Yuwei Zhong1,2#, Miaotao Zhang1,2, Yonghong Zheng1,2, Wei Peng3,4

1Department of Urology, The Sixth People’s Hospital of Huizhou, Huizhou, China; 2Department of Urology, Affiliated Huiyang Hospital of Southern Medical University, Huizhou, China; 3Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; 4Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China

Contributions: (I) Conception and design: P Li, W Peng; (II) Administrative support: W Peng; (III) Provision of study materials or patients: P Li, Y Zhong, M Zhang, Y Zheng; (IV) Collection and assembly of data: P Li, Y Zhong, M Zhang; (V) Data analysis and interpretation: P Li, Y Zhong, W Peng; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Wei Peng, PhD. Department of Urology, The First Affiliated Hospital of Anhui Medical University, Jixi Street 218, Hefei 230022, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Jixi Street 218, Hefei 230022, China. Email: pengwei0604@hotmail.com.

Background: Testicular germ cell tumor (TGCT) is a type of tumor with relatively lower incidence but being more prevalent in young men. The expression of programmed cell death ligand 1 (PD-L1) serves as a potential biomarker for predicting the survival outcomes of other tumors. Some studies discovered higher prevalence of PD-L1 in TGCT patients who achieved favorable treatment outcomes, while other studies showed lower or absent expression of PD-L1 in TGCT with the better prognosis as well. Therefore, in order to address this controversy and clarify the association between the expression of PD-L1 and pathological features and prognosis of TGCT, this meta-analysis was conducted.

Methods: A comprehensive literature search was performed using following search terms: “testis”, “testicle”, “testicular”, “cancer”, “carcinoma”, “tumor”, “neoplasm”, “programmed cell death ligand 1”, “programmed death ligand 1”, “PD-L1”, “PDL1”, “B7 homolog 1”, “B7-H1”, “B7H1” and “CD274”. Relevant studies were retrieved according to the inclusion criteria from reputable databases including PubMed, Embase, Web of Science, Cochrane Library and China National Knowledge Infrastructure (CNKI). These studies investigated the expression of PD-L1 in both tumor cells and tumor infiltrating immune cells (TIICs) in TGCT. The overall proportion of PD-L1 positivity was assessed using R programming. Pooled hazard ratio (HR) and odds ratio (OR) with corresponding 95% confidence interval (CI) were calculated using Revman software to evaluate the involvement of PD-L1 expression in TGCT. The Newcastle-Ottawa Scale (NOS) was used to evaluate the quality assessment of included studies. Sensitivity analysis and publication bias evaluation were subsequently performed.

Results: A total of eight eligible studies compromising 1,589 patients diagnosed with TGCT were finally included in this study. PD-L1 positivity was detected in 31% and 41% of TGCT patients’ tumor cells and TIICs, respectively. The pooled data demonstrated a significant association between elevated PD-L1 expression levels in TIICs and a favorable prognosis characterized by the reduced disease progression and relapse events (HR =0.21, 95% CI: 0.13–0.33). Furthermore, PD-L1+ TIICs exhibited higher prevalence rates in seminoma (OR =2.11, 95% CI: 1.57–2.84) and embryonal carcinoma (OR =6.23, 95% CI: 2.42–16.02) patients. Notably, PD-L1 expression in TIICs displayed a tendency to increase in TGCT patients with lower stages or without lymph node metastasis.

Conclusions: PD-L1 expression was observed in choriocarcinoma tumor cells, while yolk sac tumor and teratoma tumor cells exhibited lower or absent expression of PD-L1. Conversely, PD-L1 expression in TIICs was associated with seminoma and embryonal carcinoma, which was more commonly observed in TGCT patients with lower stages and better prognosis, thereby providing a theoretical foundation for the application of immunotherapy in relapsed/refractory TGCT patients.

Keywords: Programmed cell death ligand 1 (PD-L1); testicular germ cell tumor (TGCT); clinicopathology; prognosis; meta-analysis


Submitted Dec 14, 2023. Accepted for publication Jul 09, 2024. Published online Aug 21, 2024.

doi: 10.21037/tcr-23-2302


Highlight box

Key findings

• The elevated levels of programmed cell death ligand 1 (PD-L1) in tumor infiltrating immune cells (TIICs) were significantly associated with the favorable prognosis characterized by the reduced disease progression and relapse event.

• PD-L1+ TIICs exhibited higher prevalence rates in seminoma and embryonal carcinoma patients.

What is known and what is new?

• It is known that PD-L1 expression is prevalent in testicular germ cell tumor (TGCT) specimens.

• We found that PD-L1 expression in TIICs was more prevalent in seminoma and embryonal carcinoma, which indicated a better prognosis.

What is the implication, and what should change now?

• This meta-analysis represents the first comprehensive investigation of the associations between PD-L1 expression and clinicopathological characteristics as well as prognosis in patients with TGCT, which indicates the possibility of immunotherapy.

• Future research should focus on conducting large-scale studies in TGCT with compre-hensive clinical and follow-up information.


Introduction

Testicular germ cell tumor (TGCT), although accounting for approximately 1% of new male tumors annually, is the most prevalent tumor type among young men aged 15 to 40 years (1,2). Recently, the incidence among patients in their 40s is increasing (3). TGCT is derived from germ cell neoplasia in situ generated by delayed gonocytes, and it is categorized into two histological subtypes: seminoma and nonseminomatous germ cell tumor (NSGCT), consisting of embryonal carcinoma, choriocarcinoma, yolk sac tumor and teratoma (4). These subtypes can appear alone or in combination as mixed germ cell tumor (GCT). Seminoma, generally the predominant tumor type, is characterized by the elevated levels of human chorionic gonadotropin (HCG) and tends to show fewer metastatic events and a more favorable prognosis compared to NSGCT, in which alpha fetoprotein (AFP) and HCG are produced (2,5). Besides AFP, HCG and lactate dehydrogenase (LDH), genetic variants or other molecules, such as claudin 6 or programmed cell death ligand 1 (PD-L1)/programmed cell death 1 (PD-1) signaling protein, act as serum tumor biomarkers, playing pivotal roles in the diagnosis, treatment, and prognosis of TGCT (6,7).

TGCT is sensitive to radiotherapy and chemotherapy. The implementation of platinum-based chemotherapy in TGCT results in lower mortality rates compared to other tumor types, with a 5-year overall survival (OS) rate of up to 95% (8). However, despite radical orchiectomy and/or the combined treatment with radiotherapy or chemotherapy, approximately 10–15% of TGCT patients experience relapse, and salvage treatment fails to provide optimal outcomes for 50% of these individuals (9,10). Considering both the limited efficacy and the short- or long-term side effects associated with second-line and salvage therapies, alternative novel and effective treatment strategies are investigated. Compared to normal testicular tissues with macrophages as the predominant immune cell population, testicular tissues are primarily infiltrated by T cells in TGCT, thereby making immunotherapy possible (11). Recently, immunotherapy targeting immune checkpoint inhibitors such as PD-L1 or PD-1 is explored in the treatments of TGCT.

PD-1 is primarily expressed in lymphocytes and interacts with its ligand PD-L1, which can be found in tumor or immune cells. This interaction leads to the inhibition of T cell-mediated antitumor activity (12). Currently, the use of anti-PD-L1/PD-1, either alone or in combination with conventional radiotherapy or chemotherapy, demonstrates promising results in the treatments of various tumors, including lung cancer, kidney cancer, melanoma, hematologic tumors, as well as TGCT (7,13). Several case reports showed stable conditions or positive responses, such as decreased serum biomarkers and tumor volumes, in patients with relapsed/refractory TGCT following the administration of anti-PD-1 antibodies (14-16). Conversely, other evidence observed that the inhibition of PD-L1 failed to elicit antitumor effects in relapsed/refractory TGCT patients (17).

To a large extent, the effect of immunotherapy in TGCT depends on the expression of PD-L1 or PD-1. Some experimental studies revealed a higher prevalence of PD-L1 in TGCT specimens compared to normal testicular tissues, supporting the potential of immunotherapy targeting the PD-L1/PD-1 pathway in TGCT (18,19). While, other studies obtained the contradictory results showing the absent or lower expression of PD-L1 in TGCT patients (20-22). In addition, TGCT patients with higher expression of PD-L1 often achieved favorable treatment outcomes (18,19). However, other studies indicated that TGCT patients with lower PD-L1 expression had better prognosis (21). Therefore, in order to clarify the general expression of PD-L1 in TGCT, and its correlation with tumor pathology and prognosis, this meta-analysis was conducted. We present this article in accordance with the PRISMA reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2302/rc) (23).


Methods

This meta-analysis was registered on the International Platform of Registered Systematic Review and Meta-analysis Protocols (INPLASY, registration number: INPLASY2023120108). Ethical committee approval was not necessary since the analyzed data were derived from published articles. Any discrepancies between reviewers were resolved through discussion.

Search strategy

A comprehensive literature search was independently conducted by two reviewers (P.L. and W.P.) on the following databases: PubMed, Embase, Web of Science, Cochrane Library and China National Knowledge Infrastructure (CNKI), until May 6, 2024. The search terms utilized included “testis”, “testicle”, “testicular”, “cancer”, “carcinoma”, “tumor”, “neoplasm”, “programmed cell death ligand 1”, “programmed death ligand 1”, “PD-L1”, “PDL1”, “B7 homolog 1”, “B7-H1”, “B7H1” and “CD274”. For the studies included that required full-text retrieval, their references were scrutinized to identify additional relevant publications. An illustrative example of the search strategy performed in the PubMed database is shown in Appendix 1.

Eligibility criteria

Following a thorough evaluation of the title, abstract, and full-text of the retrieved literature by two reviewers (P.L. and W.P.), studies investigating the correlation between PD-L1 expression and TGCT were selected based on the Population, Intervention, Comparison, Outcomes and Study (PICOS) principal: (I) population: patients were diagnosed with TGCT; (II) intervention: PD-L1 expression in TGCT patients was assessed using immunohistochemistry; (III) comparison: the expression of PD-L1 was categorized into positive (high) and negative (low); (IV) outcomes: the study provided adequate information regarding the pathology or prognosis of TGCT patients; (V) study: cohort or case control studies.

Conversely, other factors were employed to exclude irrelevant studies, which encompassed: (I) failure to meet the inclusion criteria; (II) studies involving animal research, cell biology experiments, bioinformatic analyses, reviews, meta-analyses, comments, meeting abstracts, or case reports; (III) small sample sizes (n<15); (IV) duplicate studies or data; (V) unavailability of the full-text of the literature.

Data collection

The characteristics of the included studies were extracted independently by two reviewers (P.L. and Yuwei Zhong) and checked by another reviewer (M.Z.). Patients were divided into different subgroups depending on their specific situation. The useful data included (I) study information: authors’ name, publication year and country; (II) patient information: age, tumor type and stage, metastasis situation, International Germ Cell Cancer Collaborative Group (IGCCCG) risk classification, follow-up time and outcomes; (III) experiment information: staining method, PD-L1 antibody and cut-off value for PD-L1 positive; (IV) outcome information: total number of included patients and number of PD-L1 positive patients in each subpopulation. We collected and tabulated these data.

Quality assessment

The quality assessment of the included studies was carried out independently by two reviewers (Yuwei Zhong and Yonghong Zheng) using the Newcastle-Ottawa Scale (NOS), a tool containing eight items distributed across three domains: selection (0–4 scores), comparability (0–2 scores) and exposure or outcome (0–3 scores) (24). The NOS assigns a maximum score of 9, with studies scoring 6 or above considered to be of high quality.

Statistical analysis

The overall proportion of PD-L1 was calculated using the R package “meta” in R software version 4.3.0 for studies reporting a single proportion. To ensure appropriate data transformation, the Freeman-Tukey Double arcsine transformation method was applied (25). For statistical analysis, Review Manager 5.4 software (Revman, the Cochrane Collaboration, Oxford, UK) was utilized to compute heterogeneity, hazard ratio (HR), odds ratio (OR), and corresponding 95% confidence intervals (CIs), and generate forest plots. Heterogeneity among the studies was assessed using the Chi-squared test and I2 statistic. If the P value of the Chi-squared test was less than 0.1 or I2 exceeded 50%, significant heterogeneity was considered present, and a random-effects model was employed. Otherwise, a fixed-effects model was utilized (26). To explore the association between PD-L1 positivity and survival in TGCT patients, pooled HR with its 95% CI was determined. Additionally, the correlation between PD-L1 expression and clinicopathological characteristics in TGCT patients was analyzed using pooled OR and its 95% CI. Publication bias was assessed when the analysis included a minimum of five studies by utilizing Egger’s test, and sensitivity analysis was performed using the one-by-one elimination method through R programming (26). P<0.05 was considered statistically significant.


Results

Search results

Following the search strategy outlined above, a total of 992 articles were obtained from PubMed, Embase, Web of Science, Cochrane Library and CNKI databases. After removing duplicate studies, a preliminary screening of titles and abstracts was conducted. Non-original articles (n=427) and those not relevant to the scope of this study (n=268) were excluded. Subsequently, the full texts of the remaining studies were retrieved. Further exclusions were made based on specific criteria: utilization of duplicate patient samples (n=2), absence of dichotomization of PD-L1 expression into positive/high and negative/low groups (n=4), unavailability of analyzable data (n=2), and small sample size (n=3). Ultimately, a total of eight studies were included in this meta-analysis. The flow diagram illustrating the process of screening eligible publications for this study is presented in Figure 1.

Figure 1 The flow diagram for selecting the eligible studies. CNKI, China National Knowledge Infrastructure; PD-L1, programmed cell death ligand 1.

Study characteristics

All studies included in this meta-analysis were conducted from 2015 to 2024 in European and American countries except one study investigated Asian patients. With the exception of one study by Cierna et al., which involved specimens from nine patients with extragonadal GCT (21), the remaining studies focused on TGCT patients. The age of the patients ranged from 16 to 90 years. A total of 1,589 specimens were analyzed across these eligible studies. It is worth noting that both Cierna et al. and Chovanec et al. publications originated from the same research group. Consequently, there were 136 patients from the study by Chovanec et al. that were also included in the study conducted by Cierna et al. (21,27). Cierna et al. and Fankhauser et al. analyzed the expression of PD-L1 in tumor cells, while Chovanec et al., Sadigh et al. and Pęksa et al. examined its expression in tumor infiltrating immune cells (TIICs), such as lymphocytes and macrophages (18,21,27-29). Lobo et al., Möller et al. and Farahani et al. investigated PD-L1 expression in both tumor cells and TIICs (19,20,22). Immunohistochemistry was utilized in all these studies to determine the positivity of PD-L1, although different antibodies and cut-off values were employed. These variations contributed to the wide range of PD-L1 positive expression observed across different studies. The proportion of PD-L1 positive staining in tumor cells ranged from 0.0% to 69.6%, while in TIICs it ranged from 3.0% to 83.5%. Follow-up data on patient outcomes were available in four studies, where the prognostic value of PD-L1 expression was assessed using OS, progression-free survival (PFS), relapse-free survival (RFS), or other indicators. The characteristics of the studies included in this meta-analysis are presented in Table 1.

Table 1

Characteristics of studies included in this meta-analysis

Study Country Tumor Age, median [range], years Number of patients PD-L1 positive/high, n (%) Cut-off for positive Measurement method Antibody information Outcome Follow-up time, median [range]
Company Clone
Fankhauser 2015, (18) Switzerland Primary TGCT 33.5 [18–90] 329 229 (69.6) >5% IHC Cell Signaling Technology E1L3N NA NA
Cierna 2016, (21) Slovak Primary TGCT and extragonadal GCT 30 [17–67] 140 23 (16.4) QS ≥10 IHC Abcam EPR1161(2) PFS, OS 81.1 [0.3–235.8] months
Chovanec 2017, (27) Slovak Primary TGCT 30.6 [16–67] 240 61 (25.4)* HS ≥160 IHC Abcam EPR1161(2) PFS, OS 78.9 [0.4–293.7] months
Lobo 2019, (19) Portugal GCNIS-related TGCT 30 [25–36] 164 58 (35.4) or 137 (83.5)* >0% IHC Dako 22C3 RFS 15 years
Sadigh 2020, (28) USA Primary TGCT 35 [18–69] 77 16 (20.8)* NA IHC Cell Signaling Technology E1J2J NA 5.3 months
Pęksa 2021, (29) Poland Primary TGCT 32.9 [17–66] 180 89 (49.4)* HS ≥41 IHC Dako 22C3 EFS, CSS 31.4 months
Möller 2023, (20) Germany Primary TGCT NA 562 0 (0.0) or 411 (73.1)* >10% IHC MS Validated Antibodies MSVA–711R NA NA
Farahani 2024, (22) Iran Seminoma 39.6 33 21 (63.6) or 1 (3.0)* HS ≥15 IHC Sinabiotech NA NA NA

*, the positive staining of PD-L1 was detected in tumor infiltrating immune cells in this study. PD-L1, programmed cell death ligand 1; TGCT, testicular germ cell tumor; GCT, germ cell tumor; GCNIS, germ cell neoplasia in situ; IHC, immunohistochemistry; NA, not available; QS, quickscore; HS, histoscore; PFS, progression-free survival; OS, overall survival; RFS, relapse-free survival; EFS, event-free survival; CSS, cancer-specific survival.

Furthermore, all included studies were either cohort or case-control studies and obtained a NOS score between 6 and 7, indicating good methodological quality (Tables 2,3).

Table 2

Quality assessment of included case control studies by NOS

Study Selection Comparability Exposure Overall score
Adequacy of case definition Representativeness of the cases Selection of controls Definition of controls Comparability of cases and controls on the basis of the design or analysis Assessment of exposure Same method of ascertainment for cases and controls Non-response rate
Fankhauser 2015, (18) ★★ 7
Möller 2023, (20) ★★ 7
Farahani 2024, (22) ★★ 6

★, represents one score; ★★, represents two scores. NOS, Newcastle-Ottawa Scale.

Table 3

Quality assessment of included cohort studies by NOS

Study Selection Comparability Outcome Overall score
Representativeness of the exposed cohort Selection of the non-exposed cohort Ascertainment of exposure Outcome not present at start Comparability of cohorts on the basis of the design or analysis Assessment of outcome Adequate follow-up period Adequacy of follow-up
Cierna 2016, (21) ★★ 7
Chovanec 2017, (27) ★★ 7
Lobo 2019, (19) ★★ 7
Sadigh 2020, (28) ★★ 6
Pęksa 2021, (29) ★★ 7

★, represents one score; ★★, represents two scores. NOS, Newcastle-Ottawa Scale.

Overall proportion of positive PD-L1 expression in TGCT

PD-L1 expression has been observed in various types of cancer, including melanoma, lung cancer, gastric carcinoma, as well as TGCT (30). By pooling the data on patients with positive PD-L1 staining, we determined that 31% of TGCT patients exhibited high PD-L1 expression in tumor cells (random-effects, 95% CI: 0.06–0.65) (Figure 2A). Additionally, PD-L1 was found to be overexpressed in TIICs in 41% of TGCT patients (random-effects, 95% CI: 0.16–0.69) (Figure 2B). All the studies included in this analysis showed significant heterogeneity.

Figure 2 Forest plots of overall percentage of PD-L1 positive expression in tumor cells (A) and TIICs (B) in TGCT patients. Axis represents the proportion of PD-L1 positive expression in TGCT. CI, confidence interval; PD-L1, programmed cell death ligand 1; TIICs, tumor infiltrating immune cells; TGCT, testicular germ cell tumor.

Association of positive PD-L1 expression with prognostic implications in TGCT

Despite the inclusion of four studies that followed up with TGCT patients, two different studies examined the relationship between PD-L1 expression in tumor cells and the prognosis of TGCT. One study found that TGCT patients who exhibited a lack of PD-L1 expression or lower PD-L1 expression in the primary tumor had a better PFS compared to those with positive or higher PD-L1 expression (negative/lower verse positive/higher, HR =0.40, 95% CI: 0.16–1.01, P=0.0081) (21). While, the other study reported no significant difference of RFS among TGCT patients showing PD-L1 positive or negative tumor cells (19).

The majority of studies primarily focused on the correlation between PD-L1 expression in TIICs and the prognostic outcomes of TGCT patients, particularly in terms of disease progression or relapse events. Notably, Pęksa et al. analyzed this correlation in two specific subgroups, namely seminoma and NSGCT (29). Therefore, we segregated this study into two distinct sub-studies: (I) seminoma and (II) NSGCT. The results demonstrated that TGCT patients with PD-L1+ TIICs showed a more favorable prognosis and experienced fewer progression or relapse events compared to those with PD-L1-negative or lower TIIC expression (positive/higher verse negative/lower, HR =0.21, 95% CI: 0.13–0.33, P<0.00001) (Figure 3). No significant heterogeneity was observed. Thus, a fixed-effects model was utilized for this analysis.

Figure 3 Forest plots of HR to evaluate the correlation between PD-L1 expression and event (progression or relapse) in TGCT. SE, standard error; IV, inverse variance; CI, confidence interval; HR, hazard ratio; PD-L1, programmed cell death ligand 1; TGCT, testicular germ cell tumor.

Association of positive PD-L1 expression with clinicopathologic characteristics in TGCT

Histology

Seminoma is generally considered to be less aggressive in comparison to NSGCT (31). There was no significant difference found in the expression of PD-L1 in tumor cells of seminoma and NSGCT (Figure 4A). However, there was a significant correlation between positive PD-L1 staining in immune cells and seminoma. Among 757 seminoma patients and 466 NSGCT patients, 69% and 41% of them exhibited PD-L1 positive immune cell infiltration, respectively. The odds of PD-L1+ TIICs in seminoma were 111% higher than those in NSGCT (95% CI: 1.57–2.84) (Figure 4B). Afterwards, we analyzed the relationship between PD-L1 expression and individual subtypes of NSGCT. The pooled OR results revealed that PD-L1 overexpression in tumor cells was more prevalent in choriocarcinoma (OR =11.11, 95% CI: 4.82–25.60) (Figure 4C), while there was no advantage of PD-L1+ TIICs in choriocarcinoma (Figure 4D), as well as PD-L1 positive tumor cells in embryonal carcinoma (Figure 4E). The odds of PD-L1+ immune cell infiltration in embryonal carcinoma were significantly higher by a 5.23-fold change compared to other NSGCTs (OR =6.23, 95% CI: 2.42–16.02) (Figure 4F). In addition, lower expression of PD-L1 was commonly observed in yolk sac tumor cells (OR =0.54, 95% CI: 0.32–0.91), not in TIICs (Figure 4G,4H). Similarly, lower expression of PD-L1 was significantly associated with teratoma not only in tumor cells (OR =0.17, 95% CI: 0.05–0.54) but also in TIICs (OR =0.07, 95% CI: 0.01–0.44) (Figure 4I,4J).

Figure 4 Forest plots of OR to evaluate the correlation between PD-L1 expression and histological characteristics in TGCT. Comparison of PD-L1 expression in tumor cells (A,C,E,G,I) or TIICs (B,D,F,H,J) between seminoma and NSGCT (A,B), choriocarcinoma and other types of NSGCT (C,D), embryonal carcinoma and other types of NSGCT (E,F), yolk sac tumor and other types of NSGCT (G,H), as well as teratoma and other types of NSGCT (I,J). NSGCT, nonseminomatous germ cell tumor; M-H, Mantel-Haenszel; CI, confidence interval; OR, odds ratio; PD-L1, programmed cell death ligand 1; TGCT, testicular germ cell tumor; TIICs, tumor infiltrating immune cells.

Stages

There were two articles examining the correlation between PD-L1 expression in tumor cells and tumor-node-metastasis (TNM) or S stages in TGCT, revealing that higher stage TGCT patients had a higher rate of positive PD-L1 staining (18,21). However, upon extracting data from studies conducted by Lobo et al. and Pęksa et al. (19,29), we discovered that stage I tumors in TGCT patients showed a higher presence of PD-L1+ TIICs compared to stage II and III tumors (OR =0.70, 95% CI: 0.42–1.15) (Figure 5A). In the study by Chovanec et al. (27), they compared data from stage S0, S1 and S2 tumors with stage S3 tumors in TGCT, therefore it cannot be included in this meta-analysis. However, similar results were obtained, showing a significant association between PD-L1 positivity in TIICs and low-stage tumors but not high-stage tumors (P value =0.03) (27). Opposed to these results, the infiltration of PD-L1+ immune cells into TGCT with pT2–4 tumors demonstrated an advantage over tumors with pT1 stage, although no significant statistical differences were observed (Figure 5B). In addition, positive expression of PD-L1 in TIICs was more common in tumors that did not develop lymph node metastases or develop lymphovascular invasion (LVI), albeit without significant statistical difference (Figure 5C,5D).

Figure 5 Forest plots of OR to evaluate the correlation between PD-L1 expression in TIICs and TNM stages (A), T stages (B), lymph node metastasis (C) and LVI (D) in TGCT. M-H, Mantel-Haenszel; CI, confidence interval; LVI, lymphovascular invasion; OR, odds ratio; PD-L1, programmed cell death ligand 1; TIICs, tumor infiltrating immune cells; TNM, tumor-node-metastasis; TGCT, testicular germ cell tumor.

IGCCCG risk classification

Based on the histological subtype, metastasis, and levels of AFP, HCG and LDH, TGCT can be categorized into good, intermediate and poor outcome groups according to the IGCCCG classification. These groups correspond to approximately 96%, 89%, and 67% of the 5-year OS rate (32,33). In this study, we conducted two comparisons due to the grouping differences between Chovanec et al. who combined the good and intermediate risk groups (27), and Lobo et al. who combined the poor and intermediate risk groups (19). Both comparisons revealed that positive expression of PD-L1 on TIICs was predictive of a favorable prognosis in TGCT (Figure 6). However, we did not analyze PD-L1 expression on tumor cells due to insufficient data, as only one study addressed this aspect, which reported contrary results: higher expression of PD-L1 on tumor cells predicted a worse prognosis in TGCT patients (21).

Figure 6 Forest plots of OR to evaluate the correlation between PD-L1 expression and IGCCCG risk classification in TGCT. Comparison of PD-L1 expression in TIICs between poor and good/IM group (A) as well as poor/IM and good group (B). M-H, Mantel-Haenszel; CI, confidence interval; OR, odds ratio; PD-L1, programmed cell death ligand 1; IGCCCG, International Germ Cell Cancer Collaborative Group; TGCT, testicular germ cell tumor; TIICs, tumor infiltrating immune cells; IM, intermediate.

Sensitivity analysis and publication bias

Sensitivity analysis was conducted to evaluate following comparisons: (I) the overall proportion of positive PD-L1 expression in TGCT as shown in Figure 2; and (II) the comparison of positive PD-L1 expression in TIICs between seminoma and NSGCT as showed in Figure 4B. All comparisons included more than five studies. Upon excluding each individual study from the overall analysis, no significant differences were observed (Figure S1). Furthermore, publication bias was assessed using Egger’s test for the aforementioned studies. Results indicated no evidence of publication bias in all of these studies, as P values were greater than 0.05 in all comparisons [P values of comparison (I) were 0.45 and 0.38, respectively; P value of comparison (II) was 0.37].


Discussion

PD-L1, also known as PD-L1, is a protein that can be expressed constitutively or induced in various cells within different tissues, including the testis (34). In the physiological state, PD-L1 released by Sertoli cells, myoid peritubular cells, and germ cells directly or indirectly suppresses T cell activation and proliferation, thereby contributing to the establishment of immune privilege in the testes (35). Several studies reported that PD-L1 expression is generally lower in normal testis (18,21,36). Upregulation of PD-L1 expression is frequently observed in specific types of testicular tumors, such as GCTs and testicular lymphoma (37-39). In this meta-analysis, we included a total of eight eligible studies based on our search strategy. Although additional studies also confirmed the significant upregulation of PD-L1 in TGCT, particularly in seminoma (40-43), we were unable to incorporate them into the final meta-analysis due to the lack of dichotomous expression data of PD-L1.

Immune cell infiltration is a crucial characteristic observed during the progression of TGCT (11). Among the infiltrating cells in TGCT, macrophages, CD8+ and CD45R0+ T cells, as well as dendritic cells, constituted the major population (44,45). High levels of TIICs were associated with a worse prognosis in TGCT, indicating an increased risk of tumor relapse (46). In addition to tumor cells, PD-L1 expression was also detected in TIICs in TGCT, such as lymphocytes and macrophages (27,28). The pooled results indicated that compared to the average of 31% PD-L1 positivity on tumor cells, PD-L1 was more frequently expressed on TIICs in TGCT, with an overall overexpression rate of 41%. Notably, Möller et al. reported the absence of PD-L1 positive staining on tumor cells, while up to 73% of TIICs in TGCT can express PD-L1 (20). However, Farahani et al. showed the contradictory results, in which only 3% of TIICs in TGCT patients presented PD-L1 (22).

Moreover, different conclusions were reported regarding the association of PD-L1 expression with patient outcomes in TGCT. One study suggested that higher expression of PD-L1 in tumor cells correlated with PFS and OS rates, while another study found no significant difference in RFS rates between TGCT patients with PD-L1+ and PD-L1 tumor cells (19,21). In contrast, our comprehensive meta-analysis demonstrated that the presence of PD-L1+ TIICs was a significant predictor of favorable risk classification and prognosis, associated with reduced progression or relapse events. These findings suggest a contrasting role played by PD-L1 in tumor cells versus TIICs during the development of TGCT. One possible explanation for this discrepancy is that TGCT patients with PD-L1+ TIICs exhibit heightened sensitivity to various therapeutic interventions, resulting in more effective responses and consequently improving prognosis. Additionally, activated T cells can trigger negative feedback loops, leading to the development of adaptive immune resistance. Some TGCT patients may display limited or even absent immune cell infiltration, resulting in lower PD-L1 positivity rates in TIICs. However, these individuals may exhibit overexpression of PD-L1 in tumor cells, contributing to the divergent findings regarding PD-L1 expression in TIICs versus tumor cells. Similarly, elevated levels of PD-L1 expression in conjunction with TIICs are also associated with favorable prognostic stages in other types of tumors, including breast cancer, lung cancer or colorectal cancer (47).

The distribution of PD-L1 positive tumor cells and immune cells varies across different pathological subtypes of TGCT. Choriocarcinoma exhibited a higher prevalence of PD-L1+ staining in tumor cells, while seminoma and embryonal carcinoma displayed a higher frequency of PD-L1+ staining in TIICs. Remarkably, the levels of PD-L1+ tumor-associated macrophages were found to gradually decrease during the reprogramming process from pure seminoma to the seminoma component in mixed GCT, and subsequently to other NSGCT (43). Furthermore, irrespective of the PD-L1 expression pattern, its expression was significantly diminished in teratomas. These findings indicate that the PD-1/PD-L1 signaling pathway plays distinct roles among various subtypes of TGCT. Notably, PD-L1+ TIICs not only predicted the improved prognosis but also correlated with lower pathological grades.

The aforementioned observations strongly suggest that PD-L1 is prominently expressed in TGCT, particularly in TIICs, thereby providing a theoretical foundation and target support for the immunotherapy of TGCT. Simultaneously, the considerable variability in PD-L1 expression among patients highlights the necessity for personalized treatment approaches tailored to different individuals with TGCT. To date, immunotherapy outcomes for platinum-refractory TGCT are unsatisfactory. In a study involving fourteen TGCT patients experiencing relapse or lacking alternative options, no partial or complete responses were observed following PD-1 antibody treatment, despite only two of them were PD-L1 positive (48). Similarly, eight enrolled patients with relapsed or refractory TGCT received PD-L1 antibody immunotherapy but exhibited neither objective responses nor disease stabilization (17). However, certain case reports have described stable disease or partial response in PD-L1 positive TGCT patients following PD-1 antibody treatment (14,15). The existing clinical trial results do not definitively establish a correlation between PD-L1 expression and the efficacy of anti-PD-1/PD-L1 immunotherapy, warranting further validation through additional experiments.

In addition, the expression of PD-L1 in TIICs does not serve as a reliable individual predictor for tumor pathology and prognosis in TGCT. A study demonstrated that there was no significant difference in the 5-year event-free survival (EFS) rate between TGCT patients with PD-L1+ and PD-L1 TIICs. However, when the data of PD-L1 and V-domain Ig suppressor of T cell activation (VISTA) were combined, lower expression of PD-L1 and VISTA on TIICs significantly correlated with a worse EFS rate in TGCT (29). Furthermore, the systemic immune-inflammation index (SII) is identified as a useful tool for predicting survival outcomes in TGCT patients. Combining SII with the expression of PD-L1 in TIICs provided a more accurate prediction of TGCT patient prognosis (49,50).

However, there were some limitations in this meta-analysis. Firstly, the included studies were of insufficient quality and quantity to generate robust and accurate data. It is better to focus on conducting large-scale studies in TGCT with comprehensive clinical and follow-up information in the future research. Secondly, in terms of population ethnicity, only one study included Asian population, and other studies included patients from Europe and America. Therefore, it is essential to investigate TGCT populations from other regions such as Asia and Africa. Thirdly, there was heterogeneity in the meta-analysis due to the use of different clones of PD-L1 antibodies from various companies. The variation in antibody affinity may lead to different staining results in immunohistochemical experiments. Additionally, discrepancies in the criteria used to determine PD-L1 positivity among different studies can also impact the results. For example, Pęksa et al. considered histoscores of 41 or higher as indicative of PD-L1 positivity, whereas the research group led by Chovanec et al. required a histoscore greater than 160 (27,29).

In summary, it is important to recognize the limitations of this meta-analysis, including the inadequate quality and quantity of studies, potential bias in population ethnicity, and heterogeneity in antibody selection and PD-L1 positivity criteria. Future research should address these limitations to enhance the reliability and generalizability of findings in TGCT. Despite the aforementioned limitations, this meta-analysis represents the first comprehensive investigation of the associations between PD-L1 expression and clinicopathological characteristics as well as prognosis in patients with TGCT.


Conclusions

Our findings revealed that PD-L1 was expressed in both tumor cells and TIICs in TGCT. Specifically, PD-L1 expression was observed in choriocarcinoma tumor cells in NSGCT, while yolk sac tumor and teratoma tumor cells exhibited lower or absent expression of PD-L1. Conversely, PD-L1 expression in TIICs was associated with seminoma and embryonal carcinoma, and was more commonly observed in TGCT patients with lower stages or without lymph node metastasis. Furthermore, positive PD-L1 expression in TIICs was found to be a favorable prognostic indicator in TGCT patients, as it correlated with a reduced likelihood of disease progression or relapse.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the PRISMA reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2302/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2302/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2302/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Wagle NS, et al. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17-48. [Crossref] [PubMed]
  2. McHugh DJ, Gleeson JP, Feldman DR. Testicular cancer in 2023: Current status and recent progress. CA Cancer J Clin 2024;74:167-86. [Crossref] [PubMed]
  3. Ozaki Y, Narita T, Hatakeyama S, et al. Trends of incidence and age in adults with testicular germ cell tumors: a two-decade multicenter retrospective study. Transl Androl Urol 2023;12:161-7. [Crossref] [PubMed]
  4. Wang C, Tong L, Yao J, et al. A bibliometric analysis of testicular germ cell tumor research from 2000 to 2020. Transl Cancer Res 2021;10:3606-18. [Crossref] [PubMed]
  5. Patrikidou A, Cazzaniga W, Berney D, et al. European Association of Urology Guidelines on Testicular Cancer: 2023 Update. Eur Urol 2023;84:289-301. [Crossref] [PubMed]
  6. Islam R, Hansen A, Liesen A, et al. An update on the genetic predisposition of testicular germ cell tumors. Transl Androl Urol 2024;13:476-8. [Crossref] [PubMed]
  7. Schepisi G, Gianni C, Cursano MC, et al. Immune checkpoint inhibitors and Chimeric Antigen Receptor (CAR)-T cell therapy: Potential treatment options against Testicular Germ Cell Tumors. Front Immunol 2023;14:1118610. [Crossref] [PubMed]
  8. Lubberts S, Groot HJ, de Wit R, et al. Cardiovascular Disease in Testicular Cancer Survivors: Identification of Risk Factors and Impact on Quality of Life. J Clin Oncol 2023;41:3512-22. [Crossref] [PubMed]
  9. Urbini M, Bleve S, Schepisi G, et al. Biomarkers for Salvage Therapy in Testicular Germ Cell Tumors. Int J Mol Sci 2023;24:16872. [Crossref] [PubMed]
  10. Chovanec M, Adra N, Abu Zaid M, et al. High-dose chemotherapy for relapsed testicular germ cell tumours. Nat Rev Urol 2023;20:217-25. [Crossref] [PubMed]
  11. Islam R, Heyer J, Figura M, et al. T cells in testicular germ cell tumors: new evidence of fundamental contributions by rare subsets. Br J Cancer 2024;130:1893-903. [Crossref] [PubMed]
  12. Zhao Y, Qu Y, Hao C, et al. PD-1/PD-L1 axis in organ fibrosis. Front Immunol 2023;14:1145682. [Crossref] [PubMed]
  13. Chu X, Tian W, Wang Z, et al. Co-inhibition of TIGIT and PD-1/PD-L1 in Cancer Immunotherapy: Mechanisms and Clinical Trials. Mol Cancer 2023;22:93. [Crossref] [PubMed]
  14. Zschäbitz S, Lasitschka F, Jäger D, et al. Activity of immune checkpoint inhibition in platinum refractory germ-cell tumors. Ann Oncol 2016;27:1356-60. [Crossref] [PubMed]
  15. Zschäbitz S, Lasitschka F, Hadaschik B, et al. Response to anti-programmed cell death protein-1 antibodies in men treated for platinum refractory germ cell cancer relapsed after high-dose chemotherapy and stem cell transplantation. Eur J Cancer 2017;76:1-7. [Crossref] [PubMed]
  16. Shah S, Ward JE, Bao R, et al. Clinical Response of a Patient to Anti-PD-1 Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors. Cancer Immunol Res 2016;4:903-9. [Crossref] [PubMed]
  17. Mego M, Svetlovska D, Chovanec M, et al. Phase II study of avelumab in multiple relapsed/refractory germ cell cancer. Invest New Drugs 2019;37:748-54. [Crossref] [PubMed]
  18. Fankhauser CD, Curioni-Fontecedro A, Allmann V, et al. Frequent PD-L1 expression in testicular germ cell tumors. Br J Cancer 2015;113:411-3. [Crossref] [PubMed]
  19. Lobo J, Rodrigues Â, Guimarães R, et al. Detailed Characterization of Immune Cell Infiltrate and Expression of Immune Checkpoint Molecules PD-L1/CTLA-4 and MMR Proteins in Testicular Germ Cell Tumors Disclose Novel Disease Biomarkers. Cancers (Basel) 2019;11:1535. [Crossref] [PubMed]
  20. Möller K, Knöll M, Bady E, et al. PD-L1 expression and CD8 positive lymphocytes in human neoplasms: A tissue microarray study on 11,838 tumor samples. Cancer Biomark 2023;36:177-91. [Crossref] [PubMed]
  21. Cierna Z, Mego M, Miskovska V, et al. Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. Ann Oncol 2016;27:300-5. [Crossref] [PubMed]
  22. Farahani H, Dehghanian AR, Khademolhosseini A, et al. Frequent expression of CD45RO memory T cell marker as well as low to high expression of PD-1 and PD-L1 inhibitory molecules in seminoma and dysgerminoma. J Reprod Immunol 2024;161:104184. [Crossref] [PubMed]
  23. Page MJ, McKenzie JE, Bossuyt PM, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 2021;372: [PubMed]
  24. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol 2010;25:603-5. [Crossref] [PubMed]
  25. Barendregt JJ, Doi SA, Lee YY, et al. Meta-analysis of prevalence. J Epidemiol Community Health 2013;67:974-8. [Crossref] [PubMed]
  26. Peng W, Ou Y, Wang C, et al. The short- to midterm effectiveness of stemless prostheses compared to stemmed prostheses for patients who underwent total shoulder arthroplasty: a meta-analysis. J Orthop Surg Res 2019;14:469. [Crossref] [PubMed]
  27. Chovanec M, Cierna Z, Miskovska V, et al. Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors. Oncotarget 2017;8:21794-805. [Crossref] [PubMed]
  28. Sadigh S, Farahani SJ, Shah A, et al. Differences in PD-L1-Expressing Macrophages and Immune Microenvironment in Testicular Germ Cell Tumors. Am J Clin Pathol 2020;153:387-95. [Crossref] [PubMed]
  29. Pęksa R, Kunc M, Popęda M, et al. Combined Assessment of Immune Checkpoint Regulator VISTA on Tumor-Associated Immune Cells and Platelet-to-Lymphocyte Ratio Identifies Advanced Germ Cell Tumors with Higher Risk of Unfavorable Outcomes. Cancers (Basel) 2021;13:1750. [Crossref] [PubMed]
  30. Ren D, Hua Y, Yu B, et al. Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy. Mol Cancer 2020;19:19. [Crossref] [PubMed]
  31. Gilligan T, Lin DW, Aggarwal R, et al. Testicular Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019;17:1529-54. [Crossref] [PubMed]
  32. Fankhauser CD, Gerke TA, Roth L, et al. Pre-orchiectomy tumor marker levels should not be used for International Germ Cell Consensus Classification (IGCCCG) risk group assignment. J Cancer Res Clin Oncol 2019;145:781-5. [Crossref] [PubMed]
  33. Gillessen S, Sauvé N, Collette L, et al. Predicting Outcomes in Men With Metastatic Nonseminomatous Germ Cell Tumors (NSGCT): Results From the IGCCCG Update Consortium. J Clin Oncol 2021;39:1563-74. [Crossref] [PubMed]
  34. Ntemou E, Delgouffe E, Goossens E. Immune Checkpoint Inhibitors and Male Fertility: Should Fertility Preservation Options Be Considered before Treatment? Cancers (Basel) 2024;16:1176. [Crossref] [PubMed]
  35. Shinohara T, Yamamoto T, Morimoto H, et al. Allogeneic offspring produced by induction of PD-L1 in spermatogonial stem cells via self-renewal stimulation. Stem Cell Reports 2023;18:985-98. [Crossref] [PubMed]
  36. Wang LL, Li ZH, Hu XH, et al. The roles of the PD-1/PD-L1 pathway at immunologically privileged sites. Am J Reprod Immunol 2017; [Crossref] [PubMed]
  37. Pollari M, Brück O, Pellinen T, et al. PD-L1(+) tumor-associated macrophages and PD-1(+) tumor-infiltrating lymphocytes predict survival in primary testicular lymphoma. Haematologica 2018;103:1908-14. [Crossref] [PubMed]
  38. Kelsey R. Testicular cancer: PD-L1: a promising target for TGCTs. Nat Rev Urol 2016;13:62. [PubMed]
  39. Semaan A, Haddad FG, Eid R, et al. Immunotherapy: last bullet in platinum refractory germ cell testicular cancer. Future Oncol 2019;15:533-41. [Crossref] [PubMed]
  40. Al-Hogbani M, Duguay J, Wagner DC, et al. Expression of programmed death ligand-1 (PD-L1) in metastatic and postchemotherapy viable testicular germ cell tumors. Urol Oncol 2021;39:303.e1-8. [Crossref] [PubMed]
  41. Siska PJ, Johnpulle RAN, Zhou A, et al. Deep exploration of the immune infiltrate and outcome prediction in testicular cancer by quantitative multiplexed immunohistochemistry and gene expression profiling. Oncoimmunology 2017;6:e1305535. [Crossref] [PubMed]
  42. Jennewein L, Bartsch G, Gust K, et al. Increased tumor vascularization is associated with the amount of immune competent PD-1 positive cells in testicular germ cell tumors. Oncol Lett 2018;15:9852-60. [Crossref] [PubMed]
  43. Melotti S, Ambrosi F, Franceschini T, et al. TAMs PD-L1(+) in the reprogramming of germ cell tumors of the testis. Pathol Res Pract 2023;247:154540. [Crossref] [PubMed]
  44. Lu X, Luo Y, Nie X, et al. Single-cell multi-omics analysis of human testicular germ cell tumor reveals its molecular features and microenvironment. Nat Commun 2023;14:8462. [Crossref] [PubMed]
  45. Meng J, Gao J, Li X, et al. TIMEAS, a promising method for the stratification of testicular germ cell tumor patients with distinct immune microenvironment, clinical outcome and sensitivity to frontline therapies. Cell Oncol (Dordr) 2023;46:745-59. [Crossref] [PubMed]
  46. Wang Y, Ji C, Liu J, et al. A model based on tumor-infiltrating immune cells for predicting the relapse rates of patients with testicular germ cell tumors. Int Immunopharmacol 2020;86:106710. [Crossref] [PubMed]
  47. Zhang Y, Wu J, Zhao C, et al. Recent Advancement of PD-L1 Detection Technologies and Clinical Applications in the Era of Precision Cancer Therapy. J Cancer 2023;14:850-73. [Crossref] [PubMed]
  48. Adra N, Einhorn LH, Althouse SK, et al. Phase II trial of pembrolizumab in patients with platinum refractory germ-cell tumors: a Hoosier Cancer Research Network Study GU14-206. Ann Oncol 2018;29:209-14. [Crossref] [PubMed]
  49. Salazar-Valdivia FE, Valdez-Cornejo VA, Ulloque-Badaracco JR, et al. Systemic Immune-Inflammation Index and Mortality in Testicular Cancer: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2023;13:843. [Crossref] [PubMed]
  50. Chovanec M, Cierna Z, Miskovska V, et al. Systemic immune-inflammation index in germ-cell tumours. Br J Cancer 2018;118:831-8. [Crossref] [PubMed]
Cite this article as: Li P, Zhong Y, Zhang M, Zheng Y, Peng W. The expression of programmed cell death ligand 1 (PD-L1) involves in the clinicopathologic characteristics and prognostic implications of testicular germ cell tumor (TGCT): a systematic review and meta-analysis. Transl Cancer Res 2024;13(8):3944-3959. doi: 10.21037/tcr-23-2302

Download Citation