- Research
- Open access
- Published:
The association of PD-L1 expression status and the PD-1/PD-L1 inhibitor-related toxicity profile in non-small cell lung cancer
BMC Cancer volume 25, Article number: 799 (2025)
Abstract
Objective
While PD-L1 expression serves as a predictive biomarker for programmed cell death 1 and its ligand (PD-1/PD-L1) inhibitor efficacy in patients with non-small cell lung cancer (NSCLC), its association with treatment-related adverse events (TRAEs) has yet to be fully elucidated. This study systematically evaluated the correlation between PD-L1 expression status and TRAEs in patients with NSCLC.
Methods
We systematically searched the Cochrane Library, Embase, and PubMed databases from inception to June 30, 2024, to identify prospective clinical trials examining PD-1/PD-L1 inhibitors among NSCLC patients that reported treatment-related toxicity data stratified by PD-L1 expression.
Results
Twenty-six prospective trials (N = 5,453) were analyzed. At the 1%, 25%, and 50% PD-L1 cutoffs, PD-L1-negative patients presented significantly reduced risks of grade 3–4 TRAEs (OR = 0.37, 0.53, 0.41; 95% CI = 0.18–0.77, 0.31–0.90, 0.19–0.97; P < 0.01, 0.02, 0.04). Similarly, PD-L1-negative patients had significantly reduced risks of AEs leading to treatment discontinuation at the 1% and 25% PD-L1 cutoffs (OR = 0.25, 0.38; 95% CI = 0.08–0.76, 0.16–0.89; P = 0.01, 0.03) but not at the 50% PD-L1 cutoff (OR 0.28, 95% CI 0.07–1.12, P = 0.07). Subgroup analyses revealed elevated all-grade TRAEs with the 22C3 immunohistochemistry assay (P < 0.001), whereas first-line therapy recipients (P = 0.006) and open-label trial participants (P = 0.002) presented increased grade 3–4 TRAEs.
Conclusions
PD-L1 positivity may predict increased risks of grade 3–4 TRAEs and AEs leading to treatment discontinuation in NSCLC patients receiving PD-1/PD-L1 blockade. Furthermore, PD-L1 expression might be a useful biomarker for toxicity management in patients with NSCLC after PD-1/PD-L1 inhibitor treatment.
Background
Non-small cell lung cancer (NSCLC) poses a major public health threat worldwide due to its high incidence and mortality rates [1]. Currently, immune checkpoint inhibitors targeting programmed cell death 1 (PD-1) and its ligand (PD-L1) have become standard therapies for advanced NSCLC patients without actionable driver mutations (such as those in the EGFR or ALK genes), leading to the widespread adoption of these agents in clinical practice worldwide.
In addition to the efficacy of PD-1/PD-L1 inhibitor treatment, its toxicity profile is one primary concern among patients and is usually a deciding factor for clinicians when patients start any treatment in the clinic. PD-1/PD-L1 inhibitors may be correlated with various categories of treatment-related adverse events (TRAEs), including rash, diarrhea, pneumonitis, hypothyroidism, and hepatotoxicity [2]. Moreover, severe toxicities (e.g., grade 3–4 adverse events or adverse events leading to discontinuation) can result in debilitating or fatal outcomes, thereby profoundly impacting patients, families, and society [2, 3]. Furthermore, early detection and intervention are critical for managing these toxicities. Therefore, the identification of predictive biomarkers for PD-1/PD-L1 inhibitor-induced toxicity represents an urgent clinical challenge, as this information would enable precise risk stratification, facilitate personalized treatment optimization, and alleviate the socioeconomic burdens associated with managing TRAEs.
According to the National Comprehensive Cancer Network guidelines and multiple other guidelines (e.g., the European Society for Medical Oncology), treatment options for PD-1/PD-L1 inhibitor-based therapy are dependent on PD-L1 expression, owing to its utility as a biomarker for predicting response [4,5,6]. However, it remains unclear whether PD-L1 expression is associated with toxicity profiles, as current safety data are not stratified based on PD-L1 status. Moreover, the toxicity profile associated with PD-1/PD-L1 inhibitors is often understated during decision-making for treatment processes [7]. Therefore, we conducted this study to investigate whether PD-L1 expression is a predictive biomarker for the toxicity profile in NSCLC patients receiving PD-1/PD-L1 blockade.
Materials and methods
The current study of toxicity analyses was performed in strict accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines [8]. The present study was not registered.
Trial search strategy
On June 30, 2024, three independent researchers (Q. Z., H. H. and L-Y.OY.) systematically searched the Cochrane Library, Embase, and PubMed databases without language restrictions, and the results were confirmed by a third investigator (X–Y. H.). Supplementary Table S1 shows the details of the search strategy. We also searched for unpublished/ongoing trials at https://clinicaltrials.gov/and meeting abstracts (World Conference on Lung Cancer, American Association of Cancer Research, etc.) between January 1, 2012, and June 30, 2024. Furthermore, we manually searched the reference lists of the included articles.
Criteria for selection and exclusion
Two independent researchers (Q. Z. and H. H.) screened the studies, and the third investigator (L-Y.OY.) verified it. To reduce the risk of bias, we further excluded studies adopting PD-1/PD-L1 inhibitor-based combination therapy because different combination therapies may contribute substantially to the toxicity profile. The eligibility criteria were established based on the PICO framework., as follows: (1) participants: clinical trials prospectively enrolling NSCLC patients; (2) intervention: PD-1/PD-L1 inhibitor monotherapy; (3) control: none; and (4) outcome: the occurrence of TRAEs (events/incidence and sample size). Hence, non-prospective clinical trials, such as retrospective studies, case reports, and reviews, were excluded. When duplicate trials were identified, researchers retained the trial with the longest follow-up time and/or with the most recent follow-up.
Data extraction and assessment of risk of bias
Two researchers (P.H. and R.Y.) independently retrieved the standardized data, and a third researcher (W-X.W.) confirmed. The following data were extracted from the included studies: study name, study design, study type, tumor stage, line of therapy, median age, male sex (%), PD-1/PD-L1 inhibitor type, sample size (number of participants evaluable for toxicity), immunohistochemistry (IHC) assay, median treatment duration, PD-L1-stained cell type, and PD-L1 expression. The treatment-related toxicity profile (all-grade, grade 3–4, serious adverse events (SAEs), adverse events (AEs) leading to discontinuation, and fatal adverse events (FAEs)) was determined by the principal investigator of the original trial. We then extracted the aforementioned treatment-related toxicity profile to conduct the current study. When detailed information regarding TRAEs was not available, we sought to communicate with the corresponding author of the original trial for confirmation and identified the ambiguous interpretation as “not available (NA)”. AEs were assessed in accordance with the National Cancer Institute Common Terminology Criteria guidelines.
Two independent researchers (P.H. and R.Y.) used the 9-point Newcastle‒Ottawa Scale (NOS) [9] with eight items to assess the study quality in nonrandomized clinical trials. Each trial was classified into three main groups (selection, comparability, and outcomes for cohort studies) [9]. High-quality studies were identified as those with a score of 7 or higher. For randomized clinical trials, the abovementioned two researchers independently appraised each trial using the 7-item Cochrane Collaboration tool [10]. Any discrepancies were settled through discussion or consultation with a third researcher (X–Y. H.).
Statistical analysis
GraphPad Prism software (version 10.1.0; GraphPad Software, San Diego, CA) was used to perform all the statistical analyses, and two-tailed P values less than 0.05 were considered statistically significant. Among the different PD-L1 expression levels, the chi-square test or Fisher’s exact test (if appropriate) was used to calculate the frequencies of the toxicity profiles. When one (or more) value was zero, 0.5 was added to each value before the odds ratio (OR) and confidence interval (CI) were calculated. In trials examining the same anti-PD-1/PD-L1 drug, we evaluated the toxicity profile for different PD-L1 expression levels to study whether the toxicity was PD-L1 expression dependent. Subgroup analysis was carried out to measure the relationships of the line of therapy (2 or later line vs. first-line), study design (double-blind vs. open-label), clinical phase (phase 3 vs. phase 1/2), and IHC assay type (22 C3 vs. other) with the frequency of toxicity profiles in those trials enrolling the same PD-L1 expression and PD-1/PD-L1 inhibitor.
Results
Search results and major clinical characteristics
As shown in Fig. 1, we screened a total of 16,442 publications via both electronic databases and manual searches. After the initial screening, 133 trials were used for detailed eligibility assessment. Finally, the remaining 26 prospective studies [11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36] were identified for the final toxicity analyses.
Except for three trials [14, 22, 29], all trials were conducted at multiple centers, and the major clinical features are listed in Table 1. Three trials [21, 28, 36] were double-blind trials, and the majority of the trials (23 trials, 88.5%) [11,12,13,14,15,16,17,18,19,20, 22,23,24,25,26,27, 29,30,31,32,33,34,35] were open-label. Seventeen trials (65.4%) involved PD-1 inhibitors (fourteen with pembrolizumab [14, 17, 21,22,23, 25,26,27, 29, 31,32,33,34, 36] and three with nivolumab [11, 12, 30]), and nine trials (34.6%) involved PD-L1 inhibitors (five with durvalumab [16, 18,19,20, 35] and four with atezolizumab [13, 15, 24, 28]). All trials focused on nonresectable NSCLC patients (one trial [14] with EGFR-positive mutations), except one trial [22] that included resectable NSCLC patients (stage II/IIIA). Fifteen trials (57.7%) [12, 14, 20, 21, 24, 26,27,28,29,30,31,32, 34,35,36] were performed in the 1 st-line setting, seven trials (26.9%) [11, 17,18,19, 23, 25, 33] were performed in the 2- or later-line setting, three trials (11.5%) [13, 15, 16] were performed in the mixed-line setting (first-line and 2- or later-line setting), and the remaining trial [22] was performed in the neoadjuvant setting.
Overall, the total safety population included 5,453 patients with performance statuses between 0 and 2 and with a minimum of one dose of treatment. The number of patients tested for PD-L1 expression ranged from 5–683 per trial, and the median age of all patients enrolled ranged from 59 to 76. The percentage of male patients differed among the trials, varying from 36% to 78.6% of all safety calculations. For the assessment of PD-L1 expression, all the included trials used two different types of staining: three trials assessed PD-L1 expression in tumor and immune cells [13, 15, 24], and 23 trials assessed PD-L1 expression in tumor cells (88.5%). In total, five PD-L1 IHC assays involving 5 different PD-L1 antibodies (28–8, SP142, 73–10, SP263, and 22 C3) were adopted for the estimation of PD-L1 expression, and two studies (7.7%) [25, 35] did not include a detailed PD-L1 IHC assay or PD-L1 antibody (Table 1).
Study quality and risk bias assessment
The methodological quality of the nonrandomized clinical trials is listed in Supplementary Table S2 after one trial [25] with a conference abstract only was excluded. The NOS results revealed that all the trials were considered high-quality, and the average overall score was 7.3 (range 7–8). As summarized in Supplementary Table S3, we did not detect any major flaws in the risk of bias assessment among randomized clinical trials. However, there was generally a high risk of performance and detection bias because most trials were open-label and lacked blinded interventions.
The association between PD-L1 expression status and the frequency of grade 3–4 TRAEs
The overall frequency of grade 3–4 TRAEs was 15.9% (867 of the 5,453 evaluable patients). When a 1% cutoff value was set, patients who were PD-L1-negative had a significantly lower frequency of grade 3–4 TRAEs than did those with PD-L1-positive tumors in the nivolumab cohort (7.5% vs. 18.2%, OR 0.37, 95% CI 0.18–0.77; P < 0.01; Fig. 2A). The frequency of durvalumab-induced grade 3–4 TRAEs was significantly lower in PD-L1-negative patients than in PD-L1-positive patients at the 25% cutoff value (8.8% vs. 15.4%, OR 0.53, 95% CI 0.31–0.90; P = 0.02; Fig. 2B). Among patients receiving pembrolizumab, PD-L1-negative patients (< 50%) also experienced a lower frequency of grade 3–4 TRAEs (9.2% vs. 19.7%, OR 0.41, 95% CI 0.19–0.97, P = 0.04); however, PD-L1-negative (< 1%) patients had a significantly greater frequency of grade 3–4 TRAEs than PD-L1-positive (≥ 1%) patients did (60% vs. 16.2%, OR 7.74, 95% CI 1.57–43.4, P < 0.01; Fig. 2C).
Overall frequency of grade 3–4 TRAEs according to PD-1/PD-L1 inhibitor type and PD-L1 expression. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; SAEs, serious adverse events; TRAEs, treatment-related adverse events. ns indicates nonsignificant differences (P > 0.05), * indicates P < 0.05, and ** indicates P < 0.01
The relationship between PD-L1 expression status and the frequency of AEs leading to discontinuation
AEs leading to discontinuation were reported in 357 of the 5,054 evaluable patients, resulting in an overall incidence of 7.1%. The frequency of AEs leading to discontinuation was lower among patients who were PD-L1-negative (< 1%) than among those who were PD-L1-positive (≥ 1%) in the nivolumab cohort (2.8% vs. 10.6%, OR 0.25, 95% CI 0.08–0.76, P = 0.01; Fig. 3A). Similar results were observed in the durvalumab cohorts for patients who were PD-L1-negative (< 25%) versus PD-L1-positive (≥ 25%) (2.5% vs. 6.4%, OR 0.38, 95% CI 0.16–0.89, P = 0.03; Fig. 3B). Patients who were PD-L1-negative (< 50%) tended to have a lower incidence of AEs leading to discontinuation than those who were PD-L1-positive (≥ 50%) (3.1% vs. 10.1%, OR 0.28, 95% CI 0.07–1.12, P = 0.07; Fig. 3C).
Overall frequency of TRAEs leading to discontinuation according to PD-1/PD-L1 inhibitor type and PD-L1 expression. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; SAEs, serious adverse events; TRAEs, treatment-related adverse events. ns indicates nonsignificant differences (P > 0.05), and * indicates P < 0.05
The association between PD-L1 expression status and the frequency of all-grade TRAEs
The pooled frequency of all-grade TRAEs was 66.3% (3,581 of 5,405 evaluable patients). In the nivolumab cohort, the frequency of all-grade TRAEs did not significantly differ between patients with PD-L1 expression < 1% and those with PD-L1 expression ≥ 1% (61.3% vs. 67.8%, OR 0.75, 95% CI 0.49–1.16, P = 0.20; Fig. 4A). Similarly, when a 25% cutoff value was used to define PD-L1 expression, no significant differences were found between the positive and negative groups in the durvalumab cohort (59.1% vs. 61.1%, OR 0.92, 95% CI 0.66–1.28, P = 0.62; Fig. 4B). A similar pattern was found at the 1% cutoff value for the frequency of all-grade TRAEs in the pembrolizumab cohort (60.0% vs. 66.6%, OR 0.75, 95% CI 0.15–4.25, P = 0.75); however, a significantly lower frequency of all-grade TRAEs was observed in PD-L1-negative patients at the 50% cutoff value (59.1% vs. 71.3%, OR 0.41, 95% CI 0.24–0.70, P < 0.001, Fig. 4C).
Overall frequency of all-grade TRAEs according to PD-1/PD-L1 inhibitor type and PD-L1 expression. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; TRAEs, treatment-related adverse events. ns indicates nonsignificant differences (P > 0.05), * indicates P < 0.05, and *** indicates P < 0.001
The association between PD-L1 expression status and the frequency of SAEs
The frequency of treatment-related SAEs was 10.9% (332 of the 3,034 evaluable patients). No difference was detected among PD-L1-negative patients (< 1%) and PD-L1-positive patients (≥ 1%) in the nivolumab cohort (6.6% vs. 10.2%, OR 0.63, 95% CI 0.27–1.40, P = 0.26; Fig. 5A). Additionally, no statistically significant differences were detected in the durvalumab cohort when PD-L1 expression was used at a cutoff value of 25% (4.2% vs. 8.0%, OR 0.51, 95% CI 0.25–1.05; P = 0.06; Fig. 5B). However, with a 50% cutoff value, the frequency of pembrolizumab-related SAEs was lower among PD-L1-negative patients than among PD-L1-positive patients (3.1% vs. 16.7%, OR 0.16, 95% CI 0.04–0.60, P < 0.01; Fig. 5C).
Overall frequency of treatment-related SAEs according to PD-1/PD-L1 inhibitor type and PD-L1 expression. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; TRAEs, treatment-related adverse events. ns indicates nonsignificant differences (P > 0.05), and ** indicates P < 0.01
Relationship between PD-L1 expression status and the frequency of FAEs
The frequency of treatment-related FAEs was 0.81% (44 of the 5453 evaluable patients). The most common FAEs included respiratory distress/failure, including interstitial lung disease, and pneumonitis/pneumonia (50%, 22 of 44 patients). As depicted in Fig. 6A, PD-L1-negative and PD-L1-positive patients had a similar frequency of nivolumab-related FAEs at the 1% cutoff value (0.0% vs. 3.9%, OR 0.96, 95% CI 0.05–20.1; P = 1.0). The same result was noted among PD-L1-negative (< 25%) patients and PD-L1-positive (≥ 25%) patients in the durvalumab cohort (0.42% vs. 0.69%, OR 0.61, 95% CI 0.05–4.12, P = 1.0; Fig. 6B). For patients treated with pembrolizumab, a consistent outcome was detected in PD-L1-negative patients vs. PD-L1-positive patients at the 1% cutoff (0.0% vs. 1.5%, OR 5.95, 95% CI 0.32–110.0, P = 1.0) and the 50% cutoff value (0.0% vs. 0.78%, OR 0.88, 95% CI 0.05–16.18, P = 1.0, Fig. 6C).
Overall frequency of treatment-related FAEs according to PD-1/PD-L1 inhibitor type and PD-L1 expression. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; TRAEs, treatment-related adverse events. The ns indicates nonsignificant differences (P > 0.05)
Relationship between PD-L1-positivity and toxicity profile
We further assessed the PD-L1-positive results by performing an assessment of the included trials describing the toxicity profile at cutoff values of 1%, 5%, 25%, 50%, and 90%. However, a consistent PD-L1-dependent pattern between the PD-L1-positive and PD-L1-toxicity profiles was not noted among the different PD-L1 inhibitor types (Figs. 2, 3, 4, 5, 6 and 7).
Dose-dependent results of PD-L1 positivity and toxicity profiles in the atezolizumab cohort. Abbreviations: AEs, adverse events; FAEs, fatal adverse events; PD-L1, programmed cell death protein ligand-1; TRAEs, treatment-related adverse events. ns indicates nonsignificant differences (P > 0.05), and **** indicates P < 0.0001
Subgroup analyses
We further conducted a subgroup analysis to evaluate the associations of line therapy (first-line vs. 2 or later line), study design (open-label vs. double-blind), clinical phase (phase 3 vs. phase 1/2), and IHC assay type (22 C3 vs. other) with the frequency of toxicity profiles. This analysis was performed by including PD-L1 > 1% patients treated with pembrolizumab because of the relatively large sample size available. As shown in Table 2, we did not observe any significant differences in most subgroup analyses. Notably, the 22 C3 IHC assay revealed that patients had a greater frequency of all-grade TRAEs (P < 0.001). Compared with patients receiving 2 or later -line therapy, patients receiving first-line therapy experienced a greater frequency of grade 3–4 TRAEs (P = 0.006). Moreover, the frequency of grade 3–4 TRAEs was significantly lower in the open-label trials than in the double-blind trials (P = 0.002).
Discussion
Identifying risk factors for toxicity profiles is essential for tailoring therapeutic regimens in NSCLC patients [37]. While early-phase pembrolizumab trials and subsequent atezolizumab investigations failed to establish a significant association between PD-L1 expression and immune-related adverse events (irAEs) in NSCLC patients [13, 38], a real-world clinical study of pembrolizumab demonstrated that high PD-L1 expression independently predicts elevated irAE risk [39]. Moreover, the PD-L1 expression level has been demonstrated to be strongly associated with treatment efficacy, and patients who experience irAEs tend to have a significantly higher objective response rate [37, 40]. This situation indicates that PD-L1 expression may be correlated with irAEs, possibly because 1) high PD-L1 expression reflects a tumor microenvironment with more pronounced baseline immune suppression, which may lead to excessive immune activation after PD-1/PD-L1 blockade and trigger multiple inflammatory responses, and 2) PD-1/PD-L1 inhibitors may lead to stronger immune activation in nontarget organs with high PD-L1 expression through T-cell-mediated bystander effects, resulting in adverse reactions. However, irAEs represent only the immune effect of incorrect stimulation of the immune system on normal tissues, and TRAEs indicate the toxicity profile of therapy (including irAEs and non-irAEs). Overall, the relationship between the PD-L1 expression level and TRAEs is worthy of further exploration.
Our study, which included 26 prospective trials and 5,453 patients, is the first and largest study of the relationship between PD-L1 expression status and TRAEs in patients with NSCLC receiving PD-1/PD-L1 inhibitors. We observed that PD-L1-positive patients presented a greater than twofold increase in the risks of grade 3–4 TRAEs and treatment discontinuation due to AEs at most PD-L1 expression cutoffs. Notably, patients with PD-L1 expression < 1% had significantly more grade 3–4 TRAEs in the pembrolizumab cohort, which was contrary to our main findings. The inconsistent effect in the pembrolizumab cohort could be attributed to the small number of patients included (n = 5) and the relatively significantly higher incidence of grade 3–4 TRAEs [22]. Moreover, we did not detect a significantly lower frequency of AEs leading to discontinuation in PD-L1-negative patients at the 50% cutoff (P = 0.07), which may also have been biased with the small sample size (n = 65) included. Although variability in treatment duration may theoretically confound risk assessments of PD-1/PD-L1 inhibitors, our analysis revealed comparable or shorter median treatment durations in the high PD-L1 expression cohort, suggesting that these outcomes reflect intrinsic biological differences rather than differential exposure times. Future studies should be conducted to determine whether PD-L1-high tumors exhibit preexisting T-cell dysfunction that predisposes patients to posttreatment hyperactivation.
In fact, the multidimensional heterogeneity of PD-L1 IHC assays is an important confounding factor in evaluating the association between the expression of PD-L1 and toxicity risk [41, 42]. In our subgroup analysis, a higher frequency of all-grade TRAEs was found in patients in whom the 22 C3 IHC assay was used for PD-L1 expression assessment. A previous study evaluating the comparative performance of four PD-L1 IHC assays (22 C3, 28–8, SP142, and SP263) for detecting PD-L1 expression in tumors revealed different analytical sensitivities; thus, interchanging detection platforms and applying nonstandardized cutoff values may lead to PD-L1 status misclassification in specific patient cohorts [43]. Therefore, multi-institutional efforts to establish harmonized PD-L1 scoring protocols across different IHC platforms are needed.
A lower frequency of grade 3–4 TRAEs among patients enrolled in open-label trials than among those enrolled in double-blind trials may reflect reporting bias in open-label trials. Additionally, patients receiving first-line therapy had a greater frequency of grade 3–4 TRAEs. These patients may have a longer median treatment duration, and previous therapy (i.e., chemotherapy) may result in immune suppression [44]. For the use of PD-1/PD-L1 inhibitors in the treatment of NSCLC patients with certain genetic mutations, no study has compared the differences in safety profiles between PD-L1-negative and PD-L1-positive patients. A phase 2 trial (NCT02879994) [14] in 11 EGFR mutation-positive NSCLC patients receiving pembrolizumab reported TRAEs at rates of 65% (any grade), 9.1% (grade 3–4), and 0% (mortality); while the any-grade TRAE incidence and mortality results aligned with our findings, the incidence of grade 3–4 TRAEs in the previous study was lower than the incidence observed in our cohort (9.1% vs. 15.9%). Further multicenter, randomized phase III trials are needed to confirm this association.
The current study may have several limitations. First, we investigated the summary frequency of TRAEs on the basis of trial-level data rather than individual patient-level data; thus, the generalizability of the results may be limited. Second, patients in real-world clinical practice may experience a greater frequency of TRAEs, as the included clinical trials enrolled only patients with better performance status. Third, caution should be exercised in elucidating the results because subgroup analyses included a small number of patients. With additional evidence available, this topic needs to be further explored in future trials to support our results.
Conclusions
PD-L1-positive patients may suffer a greater frequency of grade 3–4 TRAEs and AEs leading to discontinuation than PD-L1-negative patients with NSCLC treated with PD-L1/PD-L1 inhibitors at most PD-L1 cutoff values. PD-L1 expression might be a useful biomarker for PD-1/PD-L1 inhibitor risk management in patients with NSCLC.
Data availability
All data generated or analysed during this study are included in this published article [and its supplementary information files].
Abbreviations
- CI:
-
Confidence interval
- EGFR:
-
Epidermal growth factor receptor
- FAEs:
-
Fatal adverse events
- NSCLC:
-
Non-small cell lung cancer
- OR:
-
Odds ratio
- PD-1:
-
Programmed cell death 1
- PD-L1:
-
Programmed cell death ligand 1
- IHC:
-
Immunohistochemistry
- SAEs:
-
Serious adverse events
- TRAEs:
-
Treatment-related adverse events
References
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin. 2021;71(3):209–49.
Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, Shabafrouz K, Ribi C, Cairoli A, Guex-Crosier Y, Kuntzer T, Michielin O, Peters S, Coukos G, Spertini F, Thompson JA, Obeid M. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–80.
Yang F, Shay C, Abousaud M, Tang C, Li Y, Qin Z, Saba NF, Teng Y. Patterns of toxicity burden for FDA-approved immune checkpoint inhibitors in the United States. J Exp Clin Cancer Res. 2023;42(1):4.
Liu X, Guo CY, Tou FF, Wen XM, Kuang YK, Zhu Q, Hu H. Association of PD-L1 expression status with the efficacy of PD-1/PD-L1 inhibitors and overall survival in solid tumours: A systematic review and meta-analysis. Int J Cancer. 2020;147(1):116–27.
Shen X, Zhao B. Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: meta-analysis. BMJ. 2018;362:k3529.
Kuang X, Xu R, Li J. Association of PD-L1 expression with survival benefit from PD-1/PD-L1 inhibitors in advanced cancer: Systematic review and meta-analysis of phase III randomized clinical trials. Crit Rev Oncol Hematol. 2024;198:104357.
Simons EA, Smith DE, Gao D, Camidge DR. Variation in Toxicity Reporting Methods for Early Phase Lung Cancer Treatment Trials at Oncology Conferences. J Thorac Oncol. 2020;15(9):1425–33.
Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Int J Surg. 2010;8(5):336–41.
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(9):603–5.
Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, Savović J, Schulz KF, Weeks L, Sterne JAC. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.
Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, Barlesi F, Kohlhäufl M, Arrieta O, Burgio MA, Fayette J, Lena H, Poddubskaya E, Gerber DE, Gettinger SN, Rudin CM, Rizvi N, Crinò L, Blumenschein GR Jr, Antonia SJ, Dorange C, Harbison CT, Graf Finckenstein F, Brahmer JR. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med. 2015;373(17):1627–39.
Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F, Ready N, Hiltermann TJN, Nair S, Juergens R, Peters S, Minenza E, Wrangle JM, Rodriguez-Abreu D, Borghaei H, Blumenschein GR Jr, Villaruz LC, Havel L, Krejci J, Corral Jaime J, Chang H, Geese WJ, Bhagavatheeswaran P, Chen AC, Socinski MA. First-Line Nivolumab in Stage IV or Recurrent Non-Small-Cell Lung Cancer. N Engl J Med. 2017;376(25):2415–26.
Peters S, Gettinger S, Johnson ML, Jänne PA, Garassino MC, Christoph D, Toh CK, Rizvi NA, Chaft JE, Carcereny Costa E, Patel JD, Chow LQM, Koczywas M, Ho C, Früh M, van den Heuvel M, Rothenstein J, Reck M, Paz-Ares L, Shepherd FA, Kurata T, Li Z, Qiu J, Kowanetz M, Mocci S, Shankar G, Sandler A, Felip E. Phase II Trial of Atezolizumab As First-Line or Subsequent Therapy for Patients With Programmed Death-Ligand 1-Selected Advanced Non-Small-Cell Lung Cancer (BIRCH). J Clin Oncol. 2017;35(24):2781–9.
Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, Coluzzi P, Ledezma B, Mendenhall M, Hunt J, Wolf B, Jones B, Madrigal J, Horton J, Spiegel M, Carroll J, Gukasyan J, Williams T, Sauer L, Wells C, Hardy A, Linares P, Lim C, Ma L, Adame C, Garon EB. A Phase II Study of Pembrolizumab in EGFR-Mutant, PD-L1+, Tyrosine Kinase Inhibitor Naïve Patients With Advanced NSCLC. J Thorac Oncol. 2018;13(8):1138–45.
Spigel DR, Chaft JE, Gettinger S, Chao BH, Dirix L, Schmid P, Chow LQM, Hicks RJ, Leon L, Fredrickson J, Kowanetz M, Sandler A, Funke R, Rizvi NA. FIR: Efficacy, Safety, and Biomarker Analysis of a Phase II Open-Label Study of Atezolizumab in PD-L1-Selected Patients With NSCLC. J Thorac Oncol. 2018;13(11):1733–42.
Antonia SJ, Balmanoukian A, Brahmer J, Ou SI, Hellmann MD, Kim SW, Ahn MJ, Kim DW, Gutierrez M, Liu SV, Schöffski P, Jäger D, Jamal R, Jerusalem G, Lutzky J, Nemunaitis J, Calabrò L, Weiss J, Gadgeel S, Bhosle J, Ascierto PA, Rebelatto MC, Narwal R, Liang M, Xiao F, Antal J, Abdullah S, Angra N, Gupta AK, Khleif SN, Segal NH. Clinical Activity, Tolerability, and Long-Term Follow-Up of Durvalumab in Patients With Advanced NSCLC. J Thorac Oncol. 2019;14(10):1794–806.
Nishio M, Takahashi T, Yoshioka H, Nakagawa K, Fukuhara T, Yamada K, Ichiki M, Tanaka H, Seto T, Sakai H, Kasahara K, Satouchi M, Han SR, Noguchi K, Shimamoto T, Kato T. KEYNOTE-025: Phase 1b study of pembrolizumab in Japanese patients with previously treated programmed death ligand 1-positive advanced non-small-cell lung cancer. Cancer Sci. 2019;110(3):1012–20.
Garassino MC, Cho BC, Kim JH, Mazières J, Vansteenkiste J, Lena H, Jaime JC, Gray JE, Powderly J, Chouaid C, Bidoli P, Wheatley-Price P, Park K, Soo RA, Poole L, Wadsworth C, Dennis PA, Rizvi NA. Final overall survival and safety update for durvalumab in third- or later-line advanced NSCLC: The phase II ATLANTIC study. Lung Cancer. 2020;147:137–42.
Planchard D, Reinmuth N, Orlov S, Fischer JR, Sugawara S, Mandziuk S, Marquez-Medina D, Novello S, Takeda Y, Soo R, Park K, McCleod M, Geater SL, Powell M, May R, Scheuring U, Stockman P, Kowalski D. ARCTIC: durvalumab with or without tremelimumab as third-line or later treatment of metastatic non-small-cell lung cancer. Ann Oncol. 2020;31(5):609–18.
Rizvi NA, Cho BC, Reinmuth N, Lee KH, Luft A, Ahn MJ, van den Heuvel MM, Cobo M, Vicente D, Smolin A, Moiseyenko V, Antonia SJ, Le Moulec S, Robinet G, Natale R, Schneider J, Shepherd FA, Geater SL, Garon EB, Kim ES, Goldberg SB, Nakagawa K, Raja R, Higgs BW, Boothman AM, Zhao L, Scheuring U, Stockman PK, Chand VK, Peters S. Durvalumab With or Without Tremelimumab vs Standard Chemotherapy in First-line Treatment of Metastatic Non-Small Cell Lung Cancer: The MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6(5):661–74.
Boyer M, Şendur MAN, Rodríguez-Abreu D, Park K, Lee DH, Çiçin I, Yumuk PF, Orlandi FJ, Leal TA, Molinier O, Soparattanapaisarn N, Langleben A, Califano R, Medgyasszay B, Hsia TC, Otterson GA, Xu L, Piperdi B, Samkari A, Reck M. Pembrolizumab Plus Ipilimumab or Placebo for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50%: Randomized, Double-Blind Phase III KEYNOTE-598 Study. J Clin Oncol. 2021;39(21):2327–38.
Eichhorn F, Klotz LV, Kriegsmann M, Bischoff H, Schneider MA, Muley T, Kriegsmann K, Haberkorn U, Heussel CP, Savai R, Zoernig I, Jaeger D, Thomas M, Hoffmann H, Winter H, Eichhorn ME. Neoadjuvant anti-programmed death-1 immunotherapy by pembrolizumab in resectable non-small cell lung cancer: First clinical experience. Lung Cancer. 2021;153:150–7.
Herbst RS, Garon EB, Kim DW, Cho BC, Gervais R, Perez-Gracia JL, Han JY, Majem M, Forster MD, Monnet I, Novello S, Gubens MA, Boyer M, Su WC, Samkari A, Jensen EH, Kobie J, Piperdi B, Baas P. Five Year Survival Update From KEYNOTE-010: Pembrolizumab Versus Docetaxel for Previously Treated, Programmed Death-Ligand 1-Positive Advanced NSCLC. J Thoracic Oncol. 2021;16(10):1718–32.
Jassem J, de Marinis F, Giaccone G, Vergnenegre A, Barrios CH, Morise M, Felip E, Oprean C, Kim YC, Andric Z, et al. Updated Overall Survival Analysis From IMpower110: atezolizumab Versus Platinum-Based Chemotherapy in Treatment-Naive Programmed Death-Ligand 1-Selected NSCLC. J Thorac Oncol. 2021;16(11):1872–82.
Ponce Aix S, Carcereny Costa E, Bosch-Barrera J, Felip Font E, Guirado M, Coves Sarto J, Majem Tarruella M, Juan Vidal OJ, Dalmau Portulas E, Diz P, Ortega Granados ALO, Domine Gomez M, Blasco Cordellat A, Mosquera Martinez J, Sala Gonzalez MA, Dorta M. Calvo de Juan V, Zugazagoitia J, Enguita AB, Paz-Ares L: Pembrolizumab re-challenge in patients with relapsed non-small cell lung cancer (NSCLC): A preliminary report of the REPLAY phase II trial - cohort I. Ann Oncol. 2021;32:S1450.
Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, O’Brien M, Rao S, Hotta K, Leal TA, Riess JW, Jensen E, Zhao B, Pietanza MC, Brahmer JR. Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50. J Clin Oncol. 2021;39(21):2339–49.
Spigel D, Jotte R, Nemunaitis J, Shum M, Schneider J, Goldschmidt J, Eisenstein J, Berz D, Seneviratne L, Socoteanu M, Bhanderi V, Konduri K, Xia M, Wang H, Hozak RR, Gueorguieva I, Ferry D, Gandhi L, Chao BH, Rybkin I. Randomized Phase 2 Studies of Checkpoint Inhibitors Alone or in Combination With Pegilodecakin in Patients With Metastatic NSCLC (CYPRESS 1 and CYPRESS 2). J Thoracic Oncol. 2021;16(2):327–33.
Cho BC, Abreu DR, Hussein M, Cobo M, Patel AJ, Secen N, Lee KH, Massuti B, Hiret S, Yang JCH, Barlesi F, Lee DH, Ares LP, Hsieh RW, Patil NS, Twomey P, Yang X, Meng R, Johnson ML. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022;23(6):781–92.
Lo Russo G, Sgambelluri F, Prelaj A, Galli F, Manglaviti S, Bottiglieri A, Di Mauro RM, Ferrara R, Galli G, Signorelli D, De Toma A, Occhipinti M, Brambilla M, Rulli E, Triulzi T, Torelli T, Agnelli L, Brich S, Martinetti A, Dumitrascu AD, Torri V, Pruneri G, Fabbri A, de Braud F, Anichini A, Proto C, Ganzinelli M, Mortarini R, Garassino MC. PEOPLE (NCT03447678), a first-line phase II pembrolizumab trial, in negative and low PD-L1 advanced NSCLC: clinical outcomes and association with circulating immune biomarkers. ESMO Open. 2022;7(6):100645.
Brahmer JR, Lee JS, Ciuleanu TE, Bernabe Caro R, Nishio M, Urban L, Audigier-Valette C, Lupinacci L, Sangha R, Pluzanski A, Burgers J, Mahave M, Ahmed S, Schoenfeld AJ, Paz-Ares LG, Reck M, Borghaei H, O’Byrne KJ, Gupta RG, Bushong J, Li L, Blum SI, Eccles LJ, Ramalingam SS. Five-Year Survival Outcomes With Nivolumab Plus Ipilimumab Versus Chemotherapy as First-Line Treatment for Metastatic Non-Small-Cell Lung Cancer in CheckMate 227. J Clin Oncol. 2023;41(6):1200–12.
Cho BC, Lee JS, Wu YL, Cicin I, Dols MC, Ahn MJ, Cuppens K, Veillon R, Nadal E, Dias JM, Martin C, Reck M, Garon EB, Felip E, Paz-Ares L, Mornex F, Vokes EE, Adjei AA, Robinson C, Sato M, Vugmeyster Y, Machl A, Audhuy F, Chaudhary S, Barlesi F. Bintrafusp Alfa Versus Pembrolizumab in Patients With Treatment-Naive, Programmed Death-Ligand 1-High Advanced NSCLC: A Randomized, Open-Label, Phase 3 Trial. J Thoracic Oncol. 2023;18(12):1731–42.
de Castro G, Jr., Kudaba I, Wu YL, Lopes G, Kowalski DM, Turna HZ, Caglevic C, Zhang L, Karaszewska B, Laktionov KK, Srimuninnimit V, Bondarenko I, Kubota K, Mukherjee R, Lin J, Souza F, Mok TSK, Cho BC. Five-Year Outcomes With Pembrolizumab Versus Chemotherapy as First-Line Therapy in Patients With Non-Small-Cell Lung Cancer and Programmed Death Ligand-1 Tumor Proportion Score ≥ 1% in the KEYNOTE-042 Study. J Clin Oncol. 2023;41(11):1986-1991.
Ren S, Feng J, Ma S, Chen H, Ma Z, Huang C, Zhang L, He J, Wang C, Zhou J, Danchaivijtr P, Wang CC, Vynnychenko I, Wang K, Orlandi F, Sriuranpong V, Li B, Ge J, Dang T, Zhou C. KEYNOTE-033: Randomized phase 3 study of pembrolizumab vs docetaxel in previously treated, PD-L1-positive, advanced NSCLC. Int J Cancer. 2023;153(3):623–34.
Maggie Liu S-Y, Huang J, Deng J-Y, Xu C-R, Yan H-H, Yang M-Y, Li Y-S, Ke EE, Zheng M-Y, Wang Z, Lin J-X, Gan B, Zhang X-C, Chen H-J, Wang B-C, Tu H-Y, Yang J-J, Zhong W-Z, Li Y, Zhou Q, Wu Y-L. PD-L1 expression guidance on sintilimab versus pembrolizumab with or without platinum-doublet chemotherapy in untreated patients with advanced non-small cell lung cancer (CTONG1901): A phase 2, randomized, controlled trial. Science Bulletin. 2024;69(4):535–43.
Mark M, Froesch P, Gysel K, Rothschild SI, Addeo A, Ackermann CJ, Chiquet S, Schneider M, Ribi K, Maranta AF, Bastian S, von Moos R, Joerger M, Früh M. First-line durvalumab in patients with PD-L1 positive, advanced non-small cell lung cancer (NSCLC) with a performance status of 2 (PS2) Primary analysis of the multicenter, single-arm phase II trial SAKK 19/17. Eur J cancer. 2024;200:113600.
Yang JC, Han B, De La Mora JE, Lee JS, Koralewski P, Karadurmus N, Sugawara S, Livi L, Basappa NS, Quantin X, Dudnik J, Ortiz DM, Mekhail T, Okpara CE, Dutcus C, Zimmer Z, Samkari A, Bhagwati N, Csőszi T. Pembrolizumab With or Without Lenvatinib for First-Line Metastatic NSCLC With Programmed Cell Death-Ligand 1 Tumor Proportion Score of at least 1% (LEAP-007): A Randomized, Double-Blind, Phase 3 Trial. J Thorac Oncol. 2024;19(6):941–53.
Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7(1):306.
Lisberg A, Tucker DA, Goldman JW, Wolf B, Carroll J, Hardy A, Morris K, Linares P, Adame C, Spiegel ML, Wells C, McKenzie J, Ledezma B, Mendenhall M, Abarca P, Bornazyan K, Hunt J, Moghadam N, Chong N, Nameth D, Marx C, Madrigal J, Vangala S, Shaverdian N, Elashoff D, Garon EB. Treatment-Related Adverse Events Predict Improved Clinical Outcome in NSCLC Patients on KEYNOTE-001 at a Single Center. Cancer Immunol Res. 2018;6(3):288–94.
Sugisaka J, Toi Y, Taguri M, Kawashima Y, Aiba T, Kawana S, Saito R, Aso M, Tsurumi K, Suzuki K, Shimizu H, Ono H, Domeki Y, Terayama K, Nakamura A, Yamanda S, Kimura Y, Honda Y, Sugawara S. Relationship between Programmed Cell Death Protein Ligand 1 Expression and Immune-related Adverse Events in Non-small-cell Lung Cancer Patients Treated with Pembrolizumab. Jma J. 2020;3(1):58–66.
Yoest JM. Clinical features, predictive correlates, and pathophysiology of immune-related adverse events in immune checkpoint inhibitor treatments in cancer: a short review. Immunotargets Ther. 2017;6:73–82.
Bassanelli M, Sioletic S, Martini M, Giacinti S, Viterbo A, Staddon A, Liberati F, Ceribelli A. Heterogeneity of PD-L1 Expression and Relationship with Biology of NSCLC. Anticancer Res. 2018;38(7):3789–96.
Wang Y, Zhou Y, Yang L, Lei L, He B, Cao J, Gao H. Challenges Coexist with Opportunities: Spatial Heterogeneity Expression of PD-L1 in Cancer Therapy. Adv Sci (Weinh). 2024;11(1):e2303175.
Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, Richardson W, Towne P, Hanks D, Vennapusa B, Mistry A, Kalamegham R, Averbuch S, Novotny J, Rubin E, Emancipator K, McCaffery I, Williams JA, Walker J, Longshore J, Tsao MS, Kerr KM. PD-L1 Immunohistochemistry Assays for Lung Cancer: Results from Phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project. J Thorac Oncol. 2017;12(2):208–22.
Monteran L, Ershaid N, Doron H, Zait Y. Scharff Ye, Ben-Yosef S, Avivi C, Barshack I, Sonnenblick A, Erez N: Chemotherapy-induced complement signaling modulates immunosuppression and metastatic relapse in breast cancer. Nat Commun. 2022;13(1):5797.
Acknowledgements
We are grateful to the reviewers and editors whose comments and suggestions dramatically enhanced this manuscript.
Funding
This work was supported by National Natural Science Foundation of China (No.82002872) and the Science and Technology Planning Project of General Hospital of Southern Theater Command (No. 2022 NZC002).
Author information
Authors and Affiliations
Contributions
Conceptualization: X-R. H.; Investigation: Q. Z., H. H., L-Y. OY., R. Y., W-X. W., and P. H.; Formal Analysis and Visualization: Q. Z., H. H., L-Y. OY. and X-R H; Methodology: H. H., and Q. Z.; Writing Original Draft: Q. Z., H. H., L-Y. OY., R. Y., W-X. W., P. H. and X-R H; All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Ethical approval is not applicable. This study is a study of other research studies.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Zhu, Q., Hu, H., OuYang, LY. et al. The association of PD-L1 expression status and the PD-1/PD-L1 inhibitor-related toxicity profile in non-small cell lung cancer. BMC Cancer 25, 799 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-14218-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-14218-5