DNA damage repair gene mutations predict the efficacy of platinum-based chemotherapy and immunotherapy plus platinum-based chemotherapy in advanced non-small cell lung cancer: a retrospective Chinese cohort study
Original Article

DNA damage repair gene mutations predict the efficacy of platinum-based chemotherapy and immunotherapy plus platinum-based chemotherapy in advanced non-small cell lung cancer: a retrospective Chinese cohort study

Zhiwei Xiao1#, Lingling Sun1#, Yating Zheng2, Hanrui Chen1, Xinting Zheng1, Jiamin Luo1, Chuying Gu1, Ruiting Lin3, Mengli Huang2, Yuezong Bai2, Zhe-Sheng Chen4, Connor J. Kinslow5, Jerold Loh6, Lizhu Lin1

1Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; 2The Medical Department, 3D Medicines Inc., Shanghai, China; 3The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; 4Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA; 5Department of Radiation Oncology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; 6Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore

Contributions: (I) Conception and design: Z Xiao, L Sun; (II) Administrative support: L Lin; (III) Provision of study materials or patients: L Lin, ZS Chen, Y Zheng, M Huang, Y Bai; (IV) Collection and assembly of data: H Chen, X Zheng, J Luo, C Gu, R Lin; (V) Data analysis and interpretation: Z Xiao, L Sun, Y Zheng; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work and should be considered as co-first authors.

Correspondence to: Lizhu Lin. Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Airport Road, Guangzhou 510405, China. Email: lizhulin26@yahoo.com.

Background: Platinum-based chemotherapy (PC) and immunotherapy plus platinum-based chemotherapy (IPC) remain the first-line treatment for advanced NSCLC. But only a minority patients benefit from PC, and existing biomarkers, such as PD-L1, have been shown to be defective in predicting the efficacy of IPC. Highlighting the need to identify novel biomarkers for the efficacy of PC and IPC. DNA damage repair (DDR) mutations are known to predict response to PC in solid tumors. However, the predictive value of DDR in PC and IPC of NSCLC remains unclear.

Methods: Patients diagnosed with advanced or metastatic NSCLC were retrospectively included if they underwent next generation sequencing prior to starting treatment. Primary endpoints were to explore whether DDR mutations (DDRmut) are associated with clinical outcomes of PC and IPC. Secondary end point were to explore the association between DDRmut and the choice to add immunotherapy to chemotherapy, and the impact of different DDR pathways on efficacy in PC and IPC.

Results: DDRmut showed a strong association with tumor mutation burden-high (TMB-H) versus DDR wild-type (DDRwt) and higher rates of PD-L1 TPS ≥50% positivity. In 63 patients treated with PC, ORRs were 15.38% and 2.86% for DDRmut and DDRwt subgroup (P=0.1536), and DCRs were 88.46% and 45.72% (P=0.00097) at 6 months after PC. The DDRmut patients had significantly improved median PFS (mPFS) and median overall survival (mOS) than DDRwt group (mPFS: 7.6 vs. 3.9 months, HR =1.93, 95% CI: 1.09 to 3.14, P=0.0220. mOS: 29.9 vs. 20.7 months, HR =2.31, 95% CI: 1.09 to 4.9, P=0.0250). Moreover, among 37 patients treated with IPC, ORRs were 45% and 11.76% for DDRmut and DDRwt patients (P=0.0365), and the DCRs were 95% and 70.58% (P=0.0752), respectively at 6 months after IPC. The DDRmut patients had significantly improved mPFS compared to the DDRwt group (19.5 vs. 4.5 months, HR =3.28, 95% CI: 1.53 to 9.56, P=0.0022). In DDRmut group, mPFS of IPC recipients was significantly better than that of PC recipients (19.5 vs. 7.6 months, HR =2.09, 95% CI: 0.98 to 4.42, P=0.050).

Conclusions: There is potential for DDR to serve as a positive predictor of PC and IPC in advanced NSCLC patients.

Keywords: Non-small cell lung cancer (NSCLC); platinum-based chemotherapy (PC); immunotherapy plus platinum-based chemotherapy (IPC); DNA damage repair (DDR); prediction


Submitted Aug 04, 2022. Accepted for publication Dec 19, 2022.

doi: 10.21037/tlcr-22-746


Highlight box

Key findings

• DDR genes may be a positive predictor of platinum-based chemotherapy, and immunotherapy plus platinum-based chemotherapy in patients with advanced NSCLC.

What is known and what is new?

• DDR mutations are known to predict response to platinum-based chemotherapy in some solid tumors.

• The predictive value of DDR mutations in platinum-based chemotherapy, and immunotherapy plus platinum-based chemotherapy of NSCLC. For patients with DDR mutations, median PFS receiving immunotherapy plus platinum-based chemotherapy was significantly better than those receiving platinum-based chemotherapy.

What is the implication, and what should change now?

• These results suggest that patients with DDR mutations can receive either platinum-based chemotherapy or immunotherapy plus platinum-based chemotherapy better than those with DDR wild-type, and immunotherapy plus platinum-based chemotherapy can be recommended preferentially.


Introduction

The non-small cell lung cancer (NSCLC) treatment landscape has been evolving rapidly over the past decade, with surgery, targeted therapies, immune checkpoint inhibitors (ICIs), chemotherapy, and radiation therapy as key components of disease management (1). For patients with advanced stage NSCLC, since the discovery of epidermal growth factor receptor (EGFR), driver genes including anaplastic lymphoma kinase (ALK) and ROS proto-oncogene 1 (ROS1) have been discovered successively, and targeted therapy has heralded a new era (2-4). Patients receiving targeted therapy have exhibited a significantly improved median overall survival (mOS) compared to those who did not receive targeted therapy, making it the preferred therapy for patients with advanced NSCLC who are driver gene-positive (5). Subsequently, immunotherapy became another milestone alongside targeted therapy (6). Undoubtedly, NSCLC is one of the cancer types to benefit the most from immunotherapy due to the use of programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) ICIs. However, multiple studies have shown that ICI monotherapy has excellent performance only in patients with high programmed cell death-ligand 1 (PD-L1) expression (7-9). Therefore, in driver-gene negative NSCLC with negative or low PD-L1 expression, immunotherapy plus platinum-based chemotherapy (henceforth “IPC”) is the most commonly used guideline-directed treatment, additionally, IPC is recommended even for patients with high PD-L1 levels, and immunotherapy monotherapy is recommended for those without crisis (10).

Although the degree of benefit of IPC differs based PD-L1 levels, current studies do not support the use of PD-L1 as a prognostic marker to identify patients who would not benefit from IPC. In KEYNOTE-189, in the comparison of PD-L1 TPS ≥50%, PD-L1 TPS 1–49% and PD-L1 TPS <1% subgroups, the mOS was not reached versus 21.8 months versus 17.8 months, respectively (11). Thus, it may be necessary to search for more predictive biomarkers to identify responders and non-responders with IPC. In addition, despite the rapid development of targeted therapies, the standard of care for a significant proportion of patients with driver negative is immunotherapy (or dual immunotherapy) and chemotherapy (12-15). Platinum-based chemotherapy (henceforth “PC”) is still the standard chemotherapy backbone for advanced NSCLC. In the ECOG1594 study, PC has been associated with a 19% objective response rate (ORR), a median progression-free survival (mPFS) of 3.6 months and a median overall survival (mOS) of 7.9 months in NSCLC (regardless of pathologic type or smoking status, etc.) (16). It is clear not every patient benefit to the same degree, and further delineation to decide on upfront treatment strategy will be helpful. For example, in the IPASS study, the mOS of non-smoking lung adenocarcinoma patients treated with carboplatin plus paclitaxel was 17.4 months (17). However, there is still a lack of biomarkers that can better distinguish the corresponding patients with survival benefits, and no studies have shown which groups of people may benefit the most from PC.

Overall, despite the increasing use of targeted therapies, NSCLC patients with driver gene-negative still do not benefit from these therapies. PC and IPC remain the standard first-line treatment for advanced NSCLC. However, PC has a high toxicity rate, only a small number of patients benefit from PC, there are no effective biomarkers predicting the efficacy of PC. Although immunohistochemical detection of PD-L1 expression level and the immune response to treatment of NSCLC related extensively, but the existing studies (10,11) have shown that has all PD-L1 expression level (including negative expression) of cancer patients are likely to gain long-term clinical immunotherapy, highlighting the recognition immune treatment curative effect of the necessity of new biomarkers. In addition, for patients who are unable to receive targeted therapy, biomarkers should be explored to indicate whether a patient is a better candidate for PC or IPC.

Platinum compounds exert their cytotoxic effects by forming platinum-DNA adducts that interfere with DNA repair and inhibit transcription (18). DNA-repair capacity is considered both a barrier to tumorigenesis and a crucial molecular pathway involved in resistance to PC (19). In addition, DNA damage repair (DDR) is also an emerging biomarker for immunotherapy (20). DDR gene mutations are associated with genomic instability and increased somatic tumor mutational burden (TMB), which may enhance immunogenicity by increasing tumor-specific neoantigen burden (20-23). DDR gene mutations may also enhance immune recognition and targeting through neoantigen-independent pathways (23-27). Some studies have suggested that DNA-repair capacity is both a barrier to tumorigenesis and a crucial molecular pathway involved in resistance to PC and immunotherapy (19). Inactivation of mutations of genes in DDR pathways are frequently observed in cancer. According to previous literature, there are eight DDR pathways: mismatch repair (MMR), base excision repair (BER), damage sensor (DS), Fanconi anemia (FA), homologous recombination repair (HRR), nucleotide excision repair (NER), non-homologous end-joining (NHEJ), and DNA translesion synthesis (TLS) (28). The presence of DDR mutations has been reported to correlate with improved clinical outcomes in urothelial carcinoma (29), breast cancer (30), and prostate cancer (31). Previous studies have shown that downregulation of proteins of DDR pathways is associated with worse prognosis of stage I NSCLC patients having undergone surgery, as well as with increased efficacy of PC in locally advanced or metastatic NSCLC patients (32-34), but the correlation between DDR gene mutation and platinum chemotherapy efficacy in advanced NSCLC has not been verified in clinical studies. Similarly, although previous research has shown that DDR gene mutations are associated with immunotherapy efficacy in NSCLC (35), its correlation with IPC has not been reported. In addition, with the widely application of next generation sequencing (NGS), which can detect multiple genes at the same time, studies have shown that deleterious or possibly deleterious variants of DDR genes can lead to impaired function of DDR proteins (29,36). Therefore, it is of great clinical application value to find a biomarker for predicting the efficacy of platinum chemotherapy at the DNA level. It is meaningful to explore the correlation between DDR gene mutations and the efficacy of PC and IPC in advanced NSCLC patients.

In this study, we attempted to determine whether DDR gene alterations were associated with increased sensitivity to PC and IPC among advanced NSCLC patients. We present the following article in accordance with the REMARK reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-746/rc).


Methods

Study design and patients

As shown in Figure 1, from October 2016 to September 2021, patients diagnosed with unresectable locally advanced or metastatic NSCLC and underwent tissue-based targeted exon sequencing prior to starting treatment were included from the First Affiliated Hospital of Guangzhou University of Chinese Medicine. (I) All patients were retrospectively collected and divided into two groups based on the presence of DDR pathway mutations. (II) We compared patients with DDR mutations to those without mutations in terms of genomic landscape, TMB, and PD-L1 levels. (III) After excluding patients who did not receive PC or IPC and those whose treatment was ambiguous, we also explored whether DDR mutations are associated with objective response rates (ORRs), disease control rates (DCRs) at 6 months after PC or IPC, progression-free survival (PFS), and overall survival (OS) in the PC and IPC group. (IV) We also assessed the predictive power of different DDR pathways by dividing DDR genes into different pathways and by comparing the PFS and OS of patients with specific DDR pathway mutations with those without any DDR gene mutations in the PC or IPC group.

Figure 1 Flow diagram of the study. NSCLC, non-small cell lung cancer; NGS, next-generation sequencing; DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type; PC, platinum-based chemotherapy; IPC, immunotherapy plus platinum-based chemotherapy.

In order to distinguish between DDR alterations’ prognostic role and their function as a predictor of treatment efficacy, data from untreated advanced NSCLC patients in The Cancer Genome Atlas (TCGA) database were analyzed. The TCGA dataset was accessed via cBioPortal (http://www.cbioportal.org/).

The primary endpoints of the study were to explore whether DDR mutations are associated with ORRs, DCRs, PFS, and OS of PC and IPC. The secondary end point was to explore the association between DDR mutations and the choice to add immunotherapy to chemotherapy, and the impact of different DDR pathways on efficacy in PC and IPC. An exploratory objective of this study was to identify genomic and immunologic features between patients with DDR mutations and those without mutations.

Clinical data about age, gender, Eastern Cooperative Oncology Group (ECOG) performance status, histology, smoking history, tumor blood markers, comorbidities, sites of metastatic disease, systematic treatment program, and so on, were collected from the patient medical records. Telephone follow-up and outpatient records were used for survival data, and the cutoff date for follow-up of the current study was November 2021.

The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). Collection and analysis of data were approved by the Ethics Committee of the First Affiliated Hospital of Guangzhou University of Chinese Medicine (No. K-2022-118). The requirement for informed consent was waived because patients, at the time of treatment, had consented to their anonymized medical data being analyzed and published for research purposes.

Clinical outcomes

Scans were interpreted by a dedicated chest radiologist using RECIST version 1.1 to determine ORR, DCR, and PFS (37). The PFS was calculated from the start date of PC or IPC to the date of progression, death, or last follow-up. The OS was calculated from the start date of PC to the date of death or the last follow up. At the time of last contact, patients who were still alive were examined. Patients still alive at last follow-up were censored for OS. Patients alive and without progression were censored for PFS.

Tumor tissue-based next generation sequencing

Formalin-fixed paraffin-embedded (FFPE) samples for the detection of genetic alterations were taken from needle biopsies of unresectable locally advanced or metastatic patients. All samples were independently confirmed by pathologists as consistent with the morphological characteristics of NSCLC and genomic profiling was performed in 3D Medicines Laboratory (3D Medicines Inc., Shanghai, China). The patient-generated libraries were loaded into the NovaSeq 6000 platform (Illumina, San Diego, CA, USA) for 100 bp peer sequencing with an average sequencing depth of ×1,000. The Burrows-Wheeler Aligner was used to map raw data from tumor and normal tissue paired samples to the reference human genome hg19 (BWA; version 0.7.12) (38). Picard (version 1.130; Broad Institute, Cambridge, MA, USA) was used to remove Polymerase chain reaction (PCR) repeats, and collect sequence measurements using SAMtools (version 1.1.19; http://www.htslib.org/). The variants were called only in the target area. Somatic single nucleotide variations (SNVs) were detected using an internally developed R package that detects variation based on a binomial test. Perform local rearrangement to detect inserts and deletions (indels). The variables were then filtered based on their unique support for read depth, chain bias, and base quality (39). All variants were then screened using automatic false positive tests screening pipeline to ensure sensitivity and specificity for allele frequencies ≥5%. Single nucleotide polymorphisms (SNPs) and indels were indexed with ANNOVAR (https://annovar.openbioinformatics.org/en/latest/) to note: the following database dbSNP (version 138), 1000 genomes and ESP6500 (group frequency >0.015). Only missense, stopgain, frameshift, and non-frameshift indel mutations were retained. Copy number variations (CNVs) and gene rearrangements were detected as described (39).

Targeted next-generation sequencing (NGS) was performed using a panel covering the exons of 381 cancer-related genes on an Illumina NextSeq 550 instrument (Table S1). A total of 35 genes were identified as DDR pathway genes based on searches of the PubMed, National Center for Biotechnology Information (NCBI) Gene, and NCBI BioSystems databases (Table S2).

Determination of deleterious DDR mutation status, tumor mutation burden (TMB) and PD-L1

All loss-of-function mutations in DDR genes were considered deleterious, including nonsense mutations, frameshift, or splice site alterations. To determine functional impact of missense mutations, we employed two different approaches. First, we performed an in silico functional analysis using the PolyPhen-2 (40) prediction tool to determine the functional significance of each missense mutation. Second, we reviewed all the identified missense mutations in the Catalogue of Somatic Mutations in Cancer (COSMIC) (41) and ClinVar (42) databases.

Missense mutations reported as pathogenic by COSMIC and/or ClinVar or with a PolyPhen-2 score of ≥0.95 (“probably damaging”), were classified as deleterious. Patients harboring one or more deleterious DDR mutations were defined as DDR mutations (DDRmut), and those without deleterious DDR mutations were defined as the DDR wild-type (DDRwt) subgroup.

TMB was defined as non-synonymous somatic SNVs and indels number of enzymes per megabyte that mutations in the detected coding region. All SNVs and indels in the coding region of targeted genes were considered, including missense, silent, stop gain, stop loss, in-frame, and frameshift mutations. Microsatellite instability (MSI) was assessed at 100 microsatellite loci and MSI scores for each analysis were calculated using the top 30 loci with the best coverage. An internally developed R package was used to assess the distribution of readings counting at different repeat lengths at each microsatellite site. A sample with an MSI score of at least 0.4 is considered to have high instability. Otherwise, they are considered to exhibit stability.

PD-L1 immunohistochemistry (IHC) 22C3 pharmDx assay (Agilent Technologies Inc., Santa Clara, CA, USA) or PD-L1 IHC SP263 (Roche Diagnostics, Mannheim, Baden-Wurttemberg, Germany) to evaluate PD-L1 expression in FFPE tissue slices. The staining for 22C3 was performed on the Dako Link-48 automatic staining system at Teddy Clinical Research lab (Shanghai, China) and staining for SP263 was performed on the Roche BenchMark Ultra platform at the QIAGEN Suzhou Clinical Laboratory. PD-L1 expression was measured using the tumor proportion score (TPS), which is the proportion of surviving tumor cells with partial or full membrane PD-L1 staining at any intensity. PD-L1 was positive for TPS ≥1%.

Statistical analysis

The associations between continuous variables were calculated using the Wilcoxon rank-sum test and Kruskal-Wallis test, and the χ2 test or Fisher’s exact test was used to test for associations between categorical variables. The best response was assessed based on RECIST v1.1 criteria (https://recist.eortc.org/recist-1-1-2/). The Kaplan-Meier method was used to estimate OS, PFS; the log-rank test was used to compare differences; hazard ratios (HRs) were calculated using univariate Cox regression analysis. The Cox proportional hazards regression model was used to estimate the HRs of clinicopathological factors in univariate and multivariate analyses. Trend tests were conducted using logistic regression when the outcome was binary or linear regression with log-transformation for continuous outcomes.

All P values were two-sided with statistical significance defined as P≤0.05. All statistical analyses were performed using GraphPad Prism 5 (GraphPad Software, San Diego, CA, USA) and R software version 3.6.0 (The R Foundation for Statistical Computing, Vienna, Austria).


Results

Baseline characteristics of the total NSCLC population

As shown in Figure 1, a total of 326 NSCLC patients who had undergone NGS testing were obtained from the First Affiliated Hospital of Guangzhou University of Chinese Medicine between October 2016 to September 2021. Among these cases, the median age was 61.64 years (range, 28 to 87 years), and the ratio of male to female patients was 1.9:1 (214:112). In treatment after NGS testing, 63 (19.33%) advanced cases received PC (NGS test data of patients in the PC group are shown in available online: https://cdn.amegroups.cn/static/public/tlcr-22-746-1.pdf) and 37 (11.35%) advanced cases received IPC (NGS test data of patients in the IPC group are shown in available online: https://cdn.amegroups.cn/static/public/tlcr-22-746-2.pdf); 226 (69.32%) advanced cases received other treatments. The baseline characteristics of the patients are shown in (Table 1).

Table 1

Baseline patient characteristics

Characteristics Total (n=326) DDRmut (n=202) DDRwt (n=124) P value
Age, years 0.066
   Mean (SD) 61.64 (10.80) 62.51 (10.69) 60.22 (11.23)
   Range 28–87 28–87 29–87
Gender (%) 0.058
   Female 112 (34.40) 61 (30.20) 51 (41.10)
   Male 214 (65.60) 141 (69.80) 73 (58.90)
Treatment (%) 0.471
   PC 63 (19.33) 28 (13.86) 35 (28.23)
   IPC 37 (11.35) 20 (9.90) 17 (13.71)
   Other treatments 226 (69.32) 154 (76.24) 72 (58.06)

DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild type; PC, platinum-based chemotherapy; IPC, immunotherapy plus platinum-based chemotherapy; SD, standard deviation.

Genomic landscape of the NSCLC population

In the main (326 cases) NSCLC population, TP53 had the highest mutation frequency (58.90%) followed by EGFR (35.28%) (Figure S1), which was similar to the results of previous study (43). Some 202 cases (61.96%) harboured DDRmut, with a median age of 62.51 years and the ratio of male to female patients was 2.3:1 (141:61, Table 1).

To obtain a comprehensive molecular understanding of the DDRmut cases, we investigated the genomic landscape in both DDRmut and DDRwt group patients. In the DDRmut group (202 patients), TP53 had the highest mutation frequency (62.38%) followed by EGFR (36.63%), LRP1B (18.32%), ATM (16.34%), KRAS (16.34%), and so on (Figure 2A). In the DDRwt group (124 patients), the five most frequently mutated genes were identified, including TP53 (53.23%), EGFR (33.06%), KRAS (14.52%), CDKN2A (12.10%), and LRP1B (12.10%), for which the mutation frequencies were similar but lower than those in the DDRmut group (Figure 2B). In addition, the most frequently mutated DDR genes in the DDRmut group were ATM (16.34%), BRCA2 (12.39%), PTEN (7.43%), and SMARCA4 (6.44%) (Figure S2).

Figure 2 Genomic landscape of (A) DDRmut and (B) DDRwt group patients. SNV, single nucleotide variants; CNV, copy number variations; DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type.

For the association of DDRmut and immune biomarkers, TMB was measured in 171 patients (120 in the DDRmut group and 51 in the DDRwt group), and PD-L1 was assessed in 134 patients (97 in the DDRmut group and 37 in the DDRwt group). The median TMB of the DDRmut group was significantly higher than that of the DDRwt group (7.5419 vs. 5.58659 muts/Mb, P=0.0008) (Figure 3A). The rate of strong PD-L1 (TPS ≥50%) positivity of the DDRmut group was numerically higher than that of the DDRwt group (29.03% vs. 20.00%) (Figure 3B).

Figure 3 (A) TMB and (B) PD-L1 level between DDRmut and DDRwt group. TMB, tumor mutational burden; DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type; TPS, tumor proportion score; PD-L1, programmed cell death-ligand 1.

The association of DDR mutations and outcomes after PC

The baseline characteristics of the patients undergoing PC are shown in (Table 2). In 63 patients undergoing PC, the ORRs were 15.38% for the DDRmut group and 2.86% for the DDRwt group (P=0.15358), and the DCRs were 88.46% for the DDRmut group and 45.72% for the DDRwt patients (P=0.00097) at 6 months (Figure 4A). The median PFS (mPFS) of the total population was 5.07 months (Figure S3A), which was similar to the results of previous study (mPFS: 3.6 months) (16). The DDRmut patients displayed a significantly better mPFS than the DDRwt patients [7.6 vs. 3.9 months, HR =1.93, 95% confidence interval (CI): 1.09 to 3.41, P=0.0220, Figure 4B]. The mOS of the total population was 28.2 months (Figure S3B). The DDRmut patients also displayed a significantly better mOS than the DDRwt patients (29.9 vs. 20.7 months, HR =2.31, 95% CI: 1.09 to 4.9, P=0.0250, Figure 4C).

Table 2

Baseline patient characteristics of the platinum-based chemotherapy and immunotherapy plus platinum-based chemotherapy groups

Treatment method Platinum-based chemotherapy Immunotherapy plus platinum-based chemotherapy
Total (n=63) DDRmut (n=28) DDRwt (n=35) P value Total (n=37) DDRmut (n=20) DDRwt (N=17) P value
Age, years 0.149 0.511
   Mean (SD) 59.38 (8.40) 61.11 (8.69) 58.00 (8.15) 59.38 (11.88) 60.6 (10.02) 57.94 (14.25)
   Range 43–79 43–79 43–75 30–80 32–73 30–80
Gender (%) 0.348 0.069
   Female 16 (24.65) 5 (17.90) 11 (31.40) 9 (25.60) 2 (10.00) 7 (41.20)
   Male 47 (75.35) 23 (82.10) 24 (68.60) 28 (74.40) 18 (90.00) 10 (58.80)
ECOG (%) 0.382 0.373
   1 52 (83.20) 25 (89.30) 27 (77.10) 30 (80.75) 17 (85.00) 13 (76.50)
   2 10 (15.35) 3 (10.70) 7 (20.00) 6 (16.75) 2 (10.00) 4 (23.50)
   4 1 (1.45) 0 (0.00) 1 (2.90) 1 (2.50) 1 (5.00) 0 (0.00)
Histology (%) 0.603 0.262
   LCLC 2 (3.25) 1 (3.60) 1 (2.90) 0 (0.00) 0 (0.00) 0 (0.00)
   LUAD 51 (81.40) 24 (85.70) 27 (77.10) 26 (71.20) 12 (60.00) 14 (82.40)
   LUSC 10 (15.35) 3 (10.70) 7 (20.00) 11 (28.80) 8 (40.00) 3 (17.60)
Smoke (%) 0.799 0.748
   N-Miss 2 0 2
   No 37 (60.35) 16 (57.10) 21 (63.60) 26 (69.85) 15 (75.00) 11 (64.70)
   Yes 24 (39.65) 12 (42.90) 12 (36.40) 11 (30.15) 5 (25.00) 6 (35.30)
EGFR (%) 1.000 0.482
   Wild type 48 (76.05) 21 (75.00) 27 (77.10) 33 (88.70) 19 (95.00) 14 (82.40)
   Mutation 15 (23.95) 7 (25.00) 8 (22.90) 4 (11.30) 1 (5.00) 3 (17.60)
ALK (%) 0.184 0.818
   Wild type 59 (94.30) 28 (100.00) 31 (88.60) 31 (84.10) 16 (80.00) 15 (88.20)
   Mutation 4 (5.70) 0 (0.00) 4 (11.40) 6 (15.90) 4 (20.00) 2 (11.80)
ROS1 (%) 1.000 0.720
   Wild type 57 (90.35) 25 (89.30) 32 (91.40) 33 (89.55) 17 (85.00) 16 (94.10)
   Mutation 6 (9.65) 3 (10.70) 3 (8.60) 4 (10.45) 3 (15.00) 1 (5.90)
RET (%) 1.000 1.000
   Wild type 62 (98.55) 28 (100.00) 34 (97.10) 35 (94.55) 19 (95.00) 16 (94.10)
   Mutation 1 (1.45) 0 (0.00) 1 (2.90) 2 (5.45) 1 (5.00) 1 (5.90)
CEA (%) 0.033 0.409
   N-Miss 15 5 10 3 1 2
   <5 29 (61.15) 18 (78.30) 11 (44.00) 24 (69.45) 15 (78.90) 9 (60.00)
   ≥5 19 (38.85) 5 (21.70) 14 (56.00) 10 (30.55) 4 (21.10) 6 (40.00)
CFRA21_1 (%) 1 0.211
   N-Miss 36 18 18 12 5 7
   <3.3 14 (51.45) 5 (50.00) 9 (52.90) 10 (36.65) 8 (53.30) 2 (20.00)
   ≥3.3 13 (48.55) 5 (50.00) 8 (47.10) 15 (63.35) 7 (46.70) 8 (80.00)
SCC (%) 1 1.000
   N-Miss 28 15 13 14 8 6
   <1.5 26 (74.80) 10 (76.90) 16 (72.70) 18 (78.40) 9 (75.00) 9 (81.80)
   ≥1.5 9 (25.20) 3 (23.10) 6 (27.30) 5 (21.60) 3 (25.00) 2 (18.20)
NSE (%) 0.410 0.368
   N-Miss 31 8 23 13 7 6
   <16.3 17 (55.85) 8 (66.70) 9 (45.00) 14 (59.45) 6 (46.20) 8 (72.70)
   ≥16.3 15 (44.15) 4 (33.30) 11 (55.00) 10 (40.55) 7 (53.80) 3 (27.30)
Hypertension (%) 0.299 1.000
   N-Miss 3 1 2
   No 47 (77.60) 19 (70.40) 28 (84.80) 29 (78.25) 16 (80.00) 13 (76.50)
   Yes 13 (22.40) 8 (29.60) 5 (15.20) 8 (21.75) 4 (20.00) 4 (23.50)
Digestive ulcer (%) N/A 0.934
   N-Miss 3 1 2
   No 60 (100.00) 27 (100.00) 33 (100.00) 36 (97.05) 20 (100.00) 16 (94.10)
   Yes 0 (0.00) 0 (0.00) 0 (0.00) 1 (2.95) 0 (0.00) 1 (5.90)
Cardiovascular disease (%) 0.489 1.000
   N-Miss 3 1 2
   No 54 (89.55) 23 (85.20) 31 (93.90) 32 (86.60) 17 (85.00) 15 (88.20)
   Yes 6 (10.45) 4 (14.80) 2 (6.10) 5 (13.40) 3 (15.00) 2 (11.80)
Cerebrovascular disease (%) 0.919 0.363
   N-Miss 3 1 2
   No 59 (98.15) 26 (96.30) 33 (100.00) 36 (97.30) 20 (100.00) 16 (94.10)
   Yes 1 (1.85) 1 (3.70) 0 (0.00) 1 (2.70) 0 (0.00) 1 (5.90)
Other comorbidity (%) 1.000 0.716
   No 41 (65.00) 18 (64.30) 23 (65.70) 24 (64.40) 14 (70.00) 10 (58.80)
   Yes 22 (35.00) 10 (35.70) 12 (34.30) 13 (35.60) 6 (30.00) 7 (41.20)
Visceral metastasis (%) 0.179 N/A
   N-Miss 8 1 7
   No 18 (32.55) 6 (22.20) 12 (42.90) 0 (0.00) 0 (0.00) 0 (0.00)
   Yes 37 (67.45) 21 (77.80) 16 (57.10) 37 (100.00) 20 (100.00) 17 (100.00)
Brain metastasis (%) 0.631 1.000
   N-Miss 8 1 7
   No 49 (89.05) 23 (85.20) 26 (92.90) 31 (83.70) 17 (85.00) 14 (82.40)
   Yes 6 (10.95) 4 (14.80) 2 (7.10) 6 (16.30) 3 (15.00) 3 (17.60)
Osseous metastasis (%) 0.661 0.005
   N-Miss 8 1 7
   No 47 (85.40) 22 (81.50) 25 (89.30) 19 (49.25) 15 (75.00) 4 (23.50)
   Yes 8 (14.60) 5 (18.50) 3 (10.70) 18 (50.75) 5 (25.00) 13 (76.50)
Hepatic metastasis (%) 0.985 1.000
   N-Miss 8 1 7
   No 54 (98.18) 26 (96.30) 28 (100.00) 31 (83.70) 17 (85.00) 14 (82.40)
   Yes 1 (1.82) 1 (3.70) 0 (0.00) 6 (16.30) 3 (15.00) 3 (17.60)
Lymphatic metastasis (%) 1.000 0.934
   N-Miss 8 1 7
   No 33 (60.00) 11 (91.70) 30 (81.10) 1 (2.70) 0 (0.00) 1 (5.88)
   Yes 22 (40.00) 1 (8.30) 7 (18.90) 36 (97.30) 20 (100.00) 16 (94.12)
Treatment line (%) 0.644 0.085
   1 59 (93.65) 12 (92.31) 41 (91.10) 17 (45.95) 10 (50.00) 7 (41.20)
   2 1 (1.59) 0 (0.00) 1 (2.20) 11 (29.73) 8 (40.00) 3 (17.60)
   3 2 (3.17) 1 (7.69) 2 (4.40) 7 (18.92) 1 (5.00) 6 (35.30)
   4 1 (1.59) 0 (0.00) 1 (2.20) 1 (2.70) 1 (5.00) 0 (0.00)
   5 0 (0.00) 0 (0.00) 0 (0.00) 1 (2.70) 0 (0.00) 1 (5.90)
Platinum type (%) 0.038 0.246
   Carboplatin 29 (46.03) 3 (18.75) 21 (46.67) 16 (43.24) 10 (50.00) 6 (35.30)
   Cisplatin 33 (52.38) 12 (75.00) 24 (53.33) 15 (40.54) 9 (45.00) 6 (35.30)
   Lobaplatin 1 (1.59) 1 (6.25) 0 (0.00) 6 (16.22) 1 (5.00) 5 (29.40)

DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild type; ECOG, Eastern Cooperative Oncology Group; LCLC, large cell lung cancer; LUAD, lung adenocarcinoma; LUSC, squamous cell lung carcinoma; N-Miss, the number of patients who lack this information; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; RET, RET proto-oncogene; CEA, carcinoembryonic antigen; CFRA21_1, cytokeratin 19 fragment antigen 21-1; SCC, squamous cell carcinoma antigen; NSE, neuronal-specific enolase; SD, standard deviation.

Figure 4 Efficacy and Kaplan-Meier survival curves of PC group between DDRmut and DDRwt patients. (A) ORRs and DCRs; (B) PFS in all patients receiving PC; (C) OS in all patients receiving PC; (D) PFS in patients receiving PC without carrying the driver genes; (E) OS in patients receiving PC carrying the driver genes; (F) PFS in patients receiving PC carrying driver mutations; (G) OS in in patients receiving PC carrying driver mutations. DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type; ORRs, objective response rates; DCR, disease control rates; PD, progressive disease; PR, partial response; SD, stable disease; PC, platinum-based chemotherapy; PFS, progression-free survival; OS, overall survival.

When patients carrying driver mutations in EGFR, ALK, ROS1, and RET were excluded, the mPFS of the DDRmut group was not significantly longer than that of the DDRwt patients (5 vs. 3.43 months, HR =1.72, 95% CI: 0.84 to 3.53, P=0.1300; Figure 4D), yet mOS remained significantly improved (53.4 vs. 11.5 months, HR =2.84, 95% CI: 1.09 to 7.4, P=0.0270; Figure 4E). Comparing DDRmut with DDRwt patients carrying driver mutations, there was a significant difference in mPFS (16.83 vs. 5.07 months, HR =2.73, 95% CI: 0.98 to 7.58, P=0.046; Figure 4F), but no significant differences in mOS (47.6 vs. 29.4 months, HR =1.52, 95% CI: 0.43 to 5.33, P=0.5100; Figure 4G).

In univariate analyses, deficiency in DDR genes was significantly correlated with PFS (Table 3) and OS (Table 4). In multivariate analyses, mutations in DDR genes remained a predictor of PFS and OS. Lymphatic metastases, and RET mutation were also significantly correlated with OS (Table 4). In addition, we analyzed the relationship between DDR gene mutations and the efficacy of different types of platinum agents. It was observed that the predictive effects of DDR mutations were similar for both PFS (Table 3) and OS (Table 4) between patients treated with carboplatin and cisplatin.

Table 3

Univariable and multivariable analyses for progression-free survival of platinum-based chemotherapy group

Variables Univariable analysis Multivariable analysis
HR (95% CI) P value HR (95% CI) P value
Age, years
   <60 1
   ≥60 0.84 (0.49 to 1.46) 0.545
Gender
   Female 1
   Male 1.50 (0.80 to 2.82) 0.211
ECOG
   0–1 1
   ≥2 0.93 (0.44 to 1.99) 0.860
Histology
   Non-lung adenocarcinoma 1
   Lung adenocarcinoma 1.30 (0.62 to 2.71) 0.483
Smoke
   No 1
   Yes 1.40 (0.79 to 2.48) 0.247
DDR
   Mutation 1 1.00
   Wild type 1.93 (1.09 to 3.41) 0.024 3.67 (1.53 to 8.78) 0.004
EGFR
   Mutation 1
   Wild type 1.88 (0.94 to 3.75) 0.075
ALK
   Mutation 1
   Wild type 0.84 (0.30 to 2.36) 0.747
ROS1
   Mutation 1
   Wild type 1.98 (0.77 to 5.11) 0.156
RET
   Mutation 1
   Wild type 0.33 (0.04 to 2.47) 0.279
CEA
   <5 ng/mL 1
   ≥5 ng/mL 1.71 (0.86 to 3.40) 0.128
CFRA21_1
   <3.3 ng/mL 1
   ≥3.3 ng/mL 0.81 (0.34 to 1.91) 0.634
SCC
   <1.5 ng/mL 1
   ≥1.5 ng/mL 2.59 (1.11 to 6.05) 0.028 2.41 (0.98 to 5.93) 0.055
NSE
   <16.3 ng/mL 1
   ≥16.3 ng/mL 1.42 (0.65 to 3.09) 0.376
Hypertension
   No 1
   Yes 1.39 (0.70 to 2.75) 0.349
Cardiovascular disease
   No 1
   Yes 1.73 (0.68 to 4.41) 0.254
Cerebrovascular disease
   No 1
   Yes 1.31 (0.18 to 9.63) 0.790
Other comorbidity
   No 1
   Yes 1.29 (0.70 to 2.37) 0.413
Visceral metastasis (total)
   No 1
   Yes 1.11 (0.60 to 2.05) 0.728
Brain metastasis
   No 1
   Yes 1.01 (0.40 to 2.59) 0.979
Osseous metastasis
   No 1
   Yes 0.56 (0.23 to 1.33) 0.190
Hepatic metastasis
   No 1
   Yes 0.54 (0.07 to 4.00) 0.55
Lymphatic metastasis
   No 1
   Yes 1.87 (1.04 to 3.37) 0.036 2.30 (1.01 to 5.24) 0.048
PC line
   ≤2 1
   ≥3 0.36 (0.09 to 1.52) 0.165
Platinum type
   Cisplatin 1
   Carboplatin 0.98 (0.56 to 1.71) 0.950
   Lobaplatin 0.54 (0.07 to 3.98) 0.542

ECOG, Eastern Cooperative Oncology Group; DDR, DNA damage repair; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; RET, RET proto-oncogene; CEA, carcinoembryonic antigen; CFRA21_1, cytokeratin 19 fragment antigen 21-1; SCC, squamous cell carcinoma antigen; NSE, neuronal-specific enolase; PC, platinum-based chemotherapy; HR, hazard ratio; CI, confidence interval.

Table 4

Univariable and multivariable analyses for overall survival of platinum-based chemotherapy

Variables Univariable analysis Multivariable analysis
HR (95% CI) P value HR (95% CI) P value
Age, years
   <60 1
   ≥60 1.40 (0.68 to 2.86) 0.357
Gender
   Female 1
   Male 0.96 (0.44 to 2.10) 0.917
ECOG
   0–1 1
   ≥2 0.70 (0.28 to 1.73) 0.437
Histology
   Non-lung adenocarcinoma 1
   Lung adenocarcinoma 0.36 (0.11 to 1.18) 0.091
Smoke
   No 1
   Yes 0.71 (0.33 to 1.51) 0.374
DDR
   Mutation 1
   Wild type 2.31 (1.09 to 4.90) 0.030 5.32 (2.21 to 12.83) 0.0002
EGFR
   Mutation 1
   Wild type 1.14 (0.51 to 2.55) 0.755
ALK
   Mutation 1
   Wild type 0.51 (0.18 to 1.49) 0.220
ROS1
   Mutation 1
   Wild type 1.62 (0.38 to 6.87) 0.511
RET
   Mutation 1
   Wild type 0.06 (0.01 to 0.57) 0.014 0.05 (0.00 to 0.49) 0.011
CEA
   <5 ng/mL 1
   ≥5 ng/mL 1.38 (0.64 to 2.96) 0.412
CFRA21_1
   <3.3 ng/mL 1
   ≥3.3 ng/mL 0.71 (0.23 to 2.19) 0.550
SCC
   <1.5 ng/mL 1
   ≥1.5 ng/mL 2.23 (0.86 to 5.76) 0.099 2.41 (0.98 to 5.93) 0.055
NSE
   <16.3 ng/mL 1
   ≥16.3 ng/mL 0.87 (0.34 to 2.26) 0.781
Hypertension
   No 1
   Yes 1.06 (0.46 to 2.47) 0.889
Cardiovascular disease
   No 1
   Yes 2.05 (0.70 to 5.99) 0.189
Cerebrovascular disease
   No 1
   Yes 1.17 (0.16 to 8.67) 0.880
Other comorbidity
   No 1
   Yes 1.07 (0.49 to 2.32) 0.863
Visceral metastasis (brain, osseous, hepatic, lymphatic)
   No 1
   Yes 1.16 (0.54 to 2.48) 0.709
Brain metastasis
   No 1
   Yes 1.33 (0.46 to 3.86) 0.604
Osseous metastasis
   No 1.00
   Yes 0.39 (0.11 to 1.33) 0.131
Hepatic metastasis
   No 1
   Yes 0.00 (0.00 to Inf) 0.997
Lymphatic metastasis
   No 1
   Yes 2.53 (1.22 to 5.26) 0.013 4.64 (2.02 to 10.69) 0.0003
PC line
   ≤2 1
   ≥3 0.62 (0.08 to 4.56) 0.637
Platinum type
   Cisplatin 1
   Carboplatin 0.53 (0.25 to 1.13) 0.101
   Lobaplatin 0.85 (0.11 to 6.41) 0.873

ECOG, Eastern Cooperative Oncology Group; DDR, DNA damage repair; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; RET, RET proto-oncogene; CEA, carcinoembryonic antigen; CFRA21_1, cytokeratin 19 fragment antigen 21-1; SCC, squamous cell carcinoma antigen; NSE, neuronal-specific enolase; PC, platinum-based chemotherapy; HR, hazard ratio; CI, confidence interval.

The association of DDR mutations and outcomes after IPC

In 37 patients undergoing IPC, the ORRs were 45.00% for the DDRmut group and 11.76% for the DDRwt group (P=0.03646), and the DCRs were 95.00% for the DDRmut group and 70.58% for the DDRwt group at 6 months (P=0.07523) (Figure 5A). The mPFS of the total population was 8.8 months (Figure S3C), which was similar to the results of previous study (mPFS: 8.8 months) (44). The DDRmut cases displayed a significantly better mPFS than the DDRwt patients (19.5 vs. 4.5 months, HR =3.28, 95% CI: 1.53 to 9.56, P=0.0022, Figure 5B).

Figure 5 Efficacy and Kaplan-Meier survival curves of IPC group between DDRmut and DDRwt patients. (A) ORRs and DCRs; (B) PFS in all patients receiving IPC; (C) PFS in patients receiving IPC without carrying the driver genes; (D) PFS in patients receiving IPC carrying driver mutations. DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type; ORRs, objective response rates; DCR, disease control rates; PD, progressive disease; PR, partial response; SD, stable disease; IPC, immunotherapy plus platinum-based chemotherapy; PFS, progression-free survival.

When patients carrying driver mutations in EGFR, ALK, ROS1, and RET were excluded, the mPFS of the DDRmut group was significantly longer than that of the DDRwt patients (19.5 vs. 4.5 months, HR =3.34, 95% CI: 1.1 to 10.15, P=0.0250; Figure 5C). The mPFS had no significant differences for the DDRmut patients over their wild-type counterparts in patients carrying driver mutations (NA vs. 6.5 months, HR =3.68, 95% CI: 0.72 to 18.79, P=0.0950; Figure 5D). The baseline characteristics of the patients undergoing IPC are shown in (Table 2).

In univariate analyses, DDR gene mutations were significantly correlated with PFS as well as multivariate analyses. Lymphatic metastasis was significantly correlated with PFS only in univariate analyses (Table 5). The predictive effects of DDR mutations were similar for PFS with different types of platinum agents. Patients receiving IPC in later lines (3rd line and beyond) had a worse PFS than those receiving it in earlier lines (1st to 2nd line) (Table 5).

Table 5

Univariable and multivariable analyses for progression-free survival of immunotherapy plus platinum-based chemotherapy

Variables Univariable analysis Multivariable analysis
HR (95% CI) P value HR (95% CI) P value
Age, years
   <60 1
   ≥60 1.03 (0.43 to 2.47) 0.940
Gender
   Female 1
   Male 1.27 (0.37 to 4.35) 0.703
ECOG
   0–1 1
   ≥2 0.36 (0.13 to 0.98) 0.046 0.44 (0.16 to 1.22) 0.116
Histology
   Non-lung adenocarcinoma 1
   Lung adenocarcinoma 0.92 (0.35 to 2.41) 0.864
Smoking
   No 1
   Yes 2.09 (0.85 to 5.16) 0.109
DDR
   Mutation 1
   Wild type 3.82 (1.53 to 9.56) 0.004 3 (1.14 to 7.88) 0.026
EGFR
   Mutation 1
   Wild type 1.27 (0.17 to 9.67) 0.82
ALK
   Mutation 1
   Wild type 0.84 (0.28 to 2.53) 0.753
ROS1
   Mutation 1
   Wild type 4.41 (0.58 to 33.66) 0.153
RET
   Mutation 1
   Wild type 0.00 (0.00 to Inf) 0.998
CEA
   <5 ng/mL 1
   ≥5 ng/mL 0.35 (0.08 to 1.54) 0.167
CFRA21_1
   <3.3 ng/mL 1
   ≥3.3 ng/mL 0.98 (0.33 to 2.96) 0.974
SCC
   <1.5 ng/mL 1
   ≥1.5 ng/mL 1.33 (0.27 to 6.66) 0.729
NSE
   <16.3 ng/mL 1
   ≥16.3 ng/mL 0.30 (0.06 to 1.47) 0.139
Hypertension
   No 1
   Yes 1.07 (0.35 to 3.23) 0.904
Digestive ulcer
   No 1
   Yes 4.48 (0.55 to 36.46) 0.161
Cardiovascular disease
   No 1
   Yes 0.78 (0.18 to 3.38) 0.738
Cerebrovascular disease
   No 1
   Yes 0.00 (0.00 to Inf) 0.999
Other comorbidity
   No 1
   Yes 1.20 (0.48 to 3.00) 0.693
Visceral metastasis (total)
   No 1
   Yes NA NA NA NA
Brain metastasis
   No 1
   Yes 0.79 (0.23 to 2.76) 0.712
Osseous metastasis
   No 1
   Yes 1.99 (0.83 to 4.75) 0.122
Hepatic metastasis
   No 1
   Yes 0.79 (0.23 to 2.71) 0.713
Lymphatic metastasis
   No 1
   Yes 0.05 (0.00 to 0.52) 0.013 0.12 (0.01 to 1.51) 0.102
IPC line
   ≤2 1
   ≥3 4.75 (1.73 to 13.06) 0.003 4.22 (1.38 to 12.87) 0.012
Platinum type
   Cisplatin 1
   Carboplatin 1.28 (0.49 to 3.35) 0.611
   Lobaplatin 2.18 (0.63 to 7.52) 0.217

ECOG, Eastern Cooperative Oncology Group; DDR, DNA damage repair; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; RET, RET proto-oncogene; CEA, carcinoembryonic antigen; CFRA21_1, cytokeratin 19 fragment antigen 21-1; SCC, squamous cell carcinoma antigen; NSE, neuronal-specific enolase; IPC, immunotherapy plus platinum-based chemotherapy; HR, hazard ratio; CI, confidence interval.

The association of DDR mutations and the choice to add immunotherapy to chemotherapy

In order to better identify patients who would benefit from the addition of immunotherapy, we compared the correlation between DDR mutations and outcomes on IPC and PC. For cases in the DDRmut group, the mPFS of IPC was 19.5 months, which was significantly better than that of PC (7.6 months, HR =2.09, 95% CI: 0.98 to 4.42, P=0.0500, Figure 6A). However, the mPFS of IPC and PC was not significantly different in the DDRwt group (4.5 vs. 3.9 months, HR =1.14, 95% CI: 0.58 to 2.24, P=0.7100) (Figure 6B).

Figure 6 Kaplan-Meier survival curves of PFS between PC and IPC group. (A) PFS in all DDRmut patients; (B) PFS in all DDRwt patients; (C) PFS in DDRmut patients without carrying the driver genes; (D) PFS in all DDRwt patients without carrying the driver genes; (E) PFS in DDRmut patients carrying the driver genes; (F) PFS in all DDRwt patients carrying the driver genes. IPC, immunotherapy plus platinum-based chemotherapy; PC, platinum-based chemotherapy; PFS, progression-free survival; DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type.

Similarly, when patients carrying driver mutations in EGFR, ALK, ROS1, and RET were excluded, the mPFS of those receiving IPC was significantly longer than that of those receiving PC in the DDRmut group (19.5 vs. 5 months, HR =2.6, 95% CI: 1.01 to 6.71, P=0.0400; Figure 6C). However, there were no significant differences in mPFS between the cases undergoing either IPC or PC in DDRwt group (4.5 vs. 3.43 months, HR =1.48, 95% CI: 0.64 to 3.43, P=0.3500; Figure 6D). Among patients carrying driver mutations, the mPFS was not significantly different between IPC and PC in both the DDRmut (16.8 vs. NA months, HR =1.7, 95% CI: 0.42 to 6.83, P=0.4500, Figure 6E) and DDRwt (6.5 vs. 5.07 months, HR =0.95, 95% CI: 0.26 to 3.52, P=0.9400, Figure 6F) groups. These findings suggest IPC mainly benefits patients who are driver oncogene negative, and DDRmut.

Impact of different DDR pathways on efficacy in PC and IPC

To further explore the correlation between DDRmut and efficacy, DDRmut patients were divided into three categories according to the pathway of genes, including HRR single pathway mutations (HRR), HRR combined with other pathway mutations (HRR comutations), and non-HRR pathway mutations (others), since HRR pathway gene mutations accounted for the highest proportion in DDR pathways.

The results showed that the alterations of some DDR pathways showed better efficacy. Efficacy of PC were more pronounced with patients with mutations in the HRR single pathway (HR =0.27; 95% CI: 0.06 to 1.18, P=0.064, Figure 7A) and non-HRR pathway (HR =0.37; 95% CI: 0.14 to 1.01, P=0.043, Figure 7A) for OS, and non-HRR pathway (HR =0.44; 95% CI: 0.22 to 0.91, P=0.024, Figure 7B) in PFS. For patients who received IPC, a PFS benefit was most pronounced for patients with mutations in HRR combined with other pathways (HR =0.16; 95% CI: 0.03 to 0.74, P=0.0085) (Figure 7C).

Figure 7 Influence of mutations in different DDR pathways. (A) OS in PC group; (B) PFS in PC group; (C) PFS in IPC group. OS, overall survival; PFS, progression-free survival; HRR, HRR single pathway mutations; HRR comutations, HRR combined with other pathway mutations; others, non-HRR pathway mutations; DDR, DNA damage repair; PC, platinum-based chemotherapy; IPC, immunotherapy plus platinum-based chemotherapy; DDRmut, DNA damage repair mutations; DDRwt, DNA damage repair wild-type; HRR, homologous recombination repair.

The prognostic role of DDR alterations

We evaluated whether DDR status was a prognostic factor using the survival data and sequencing data of previously untreated NSCLC patients in the TCGA database. There was no significant difference in mOS between the DDRmut and DDRwt groups in either untreated stage I patients (DDRmut vs. DDRwt =86.1 vs. 75.7 months, HR =0.97; 95% CI: 0.67 to 1.41, P=0.8800; Figure S4A) or untreated stage II-IV patients (DDRmut vs. DDRwt =27.2 vs. 32.0 months, HR =0.81; 95% CI: 0.57 to 1.16, P=0.2500; Figure S4B). These results suggested that DDR alteration status was not a prognostic factor for untreated NSCLC.


Discussion

We examined the association between DDR mutations and clinical outcomes in two cohorts of NSCLC patients treated with PC and IPC respectively. We observed that 61.96% of tumors harbored alterations in DDR genes and that the presence of DDR gene variations was associated with improved ORRs, DCRs, PFS, and OS in NSCLC patients received PC or IPC. In the comparison of the efficacy of PC and IPC, patients with DDR mutations had a better efficacy of IPC, especially those without driver gene mutations. We also demonstrated that alterations of different DDR pathways had different influences on PFS and OS on PC and IPC.

DDR gene alterations are common in NSCLC but are poorly characterized. Polymorphisms in various DDR genes, such as BRCA2 and MLH3, have been shown to be associated with leptomeningeal metastasis of NSCLC and associated with poor prognosis (45). To our knowledge, this is the first study to demonstrate an independent association between DDR gene mutations and clinical benefit to PC in patients with advanced NSCLC, the results show that DDRmut patients displayed significantly better clinical outcomes than the DDRwt patients. Similarly, Li et al. reported that advanced NSCLC patients with downregulation of APE1, or TUBB3 protein benefited from platinum plus paclitaxel chemotherapy (33). In a study on unresectable locally advanced or metastatic NSCLC patients treated with cisplatin-based chemotherapy, patients with deficient expression of BRCA1 protein had a significantly higher survival rate than those with intact expression (34). A similar phenomenon was observed in our study cohort, where alteration of the BRCA1 gene was significantly associated with better clinical outcomes (data not shown). However, a prospective study showed that ERCC1 protein expression did not predict OS or PFS for NSCLC (46). The results suggested that IHC detection of ERCC1 protein expression is not a good predictor of PC response, possibly because the reliability of IHC results was related to the quality of antibodies and subjective criteria for interpretation. On the contrary, the analysis of DDR genes using NGS can not only achieve high-throughput sequencing, but also ensure that the detection results are not affected by subjective interpretation factors of experimenters and can effectively distinguish deleterious and nondeleterious DDR gene mutations. In fact, DDR gene mutations have been shown to be associated with improved therapeutic sensitivity to PC, PARP inhibitors, ICIs, and other agents across multiple solid tumor types (29,47-49).

As early as 2018, Teo et al. reported the association between DDR gene and PD-L1 inhibitors monotherapy in advanced urethral carcinoma (50). A similar report had been made of NSCLC: DDR mutation patients had a significantly better ORRs, PFS, and OS than DDRwt with PD-L1 inhibitors monotherapy (51). Although the results of these studies indicate that patients with DDRmut can benefit from immunotherapy, patients in these studies were generally treated with immune monotherapy. As we know, immune monotherapy is recommended for patients whose PD-L1 expression exceeds 50% in NSCLC, so its clinical application is limited (7,8,52). In addition, PD-L1 combined with CTLA-4 inhibitor is still not a clinical preferred option for NSCLC treatment due to cost and efficacy limitations (52,53). Currently, IPC is the most widely used immune-related therapy in NSCLC, but its association with DDR has not been reported. This is also the first study to demonstrate an independent association between DDR gene mutations and clinical benefit to IPC in patients with advanced NSCLC, and the results show that DDRmut patients still displayed significantly better clinical outcomes than the DDRwt patients. To further explore the correlation between DDR and curative effect, we divided the DDR pathway into smaller pathways. We discovered that the most pronounced PFS benefit for IPC patients was seen in those with mutations in HRR combined with another pathway. This is similar to the results of Wang et al.’s study on the prediction of immune efficacy by mutations in HRR-MMR and HRR-BER pathways, respectively (54).

The main mechanism of the DDR pathway is the timely repair of errors during DNA replication and transcription, such as PARP involved in BER and the BRCA1/2 gene involved in HRR (47). Platinum compounds exert their cytotoxic effects by forming platinum-DNA adducts that interfere with DNA repair and inhibit transcription (55). Generally, when platinum compounds cause the platinum intrastrand crosslinks that forms on DNA, DDR genes can repair these DNA damages to a certain extent. However, when DDR genes are mutated, the DDR pathway will be blocked, which can promote the apoptosis of tumor cells (56). For example, because BRCA1/2 play key roles in HRR of DNA double-strand breaks, cancers with BRCA1/2 alterations, often have a better response to DNA cross-linking agents such as platinum compounds (57). Similarly, in our cohort, alterations in the DDR genes were significantly associated with better clinical outcomes of PC. In addition, because altering DNA damage responses mediated by exposure to cytotoxic agents or loss of normal DNA repair ability may contribute to antitumor immunity mediated by STING pathways, the DDR mutation may be more sensitive to immunotherapy (25,58-61). When cGAMP synthase (cGAS) interacts with cell-soluble DNA and catalyzes the synthesis of cGAMP, the STING pathway is activated. Activation of STING pathways in antigen-presenting cells (APCs) in tumor microenvironment drives T cells to stimulate tumor-associated antigens and promote the occurrence of anti-tumor immunity (20,62,63). In addition, chemotherapy may promote tumor immunity in two main ways, including inducing immunogenic cell death as part of its intended therapeutic effect and destroying strategies used by tumors to evade immune responses (64). Therefore, the benefits of immunotherapy and synergy of IPC may be more significant for DDRmut patients. However, strictly speaking, the mechanism by which DDR mutation enhances the sensitivity has not been clarified, neither in PC or IPC, and more mechanism studies are needed for elucidation.

This retrospective study had several limitations: (I) the retrospective design of the study and the small sample size of the PC and IPC group may have led to bias in the clinical outcomes observed; (II) more than 200 DDR genes have been reported to date (65), but only 35 were covered by the targeted panel used for this study, so it is reasonable to speculate that some DDR mutations might have been missed during detection; (III) the COSMIC and ClinVar databases are dynamic, and the degree of functional validation that underlies virulence annotations in these databases is variable; (IV) unlike previous research (29), due to the small sample size, our further division of DDR pathway was not sufficiently comprehensive; and (V) since more than half (27/37, 54.05%) of the patients in the IPC group received IPC after second-line treatment (including second-line treatment), by the time the article was submitted, only nine out of 37 patients in the IPC cohort had died, while the remaining 28 patients had not died. In addition, 15 of the 28 patients who did not have a death event were followed for less than 1 year after receiving IPC, so we believe that the use of current follow-up data to calculate OS is biased.


Conclusions

This study revealed that DDR mutations are common in NSCLC and may predict sensitivity to PC and IPC, especially in the latter. More prospective studies with larger sample sizes are needed to independently verify these findings and allow more robust analyses of individual DDR genes or gene subsets. Further research into the underlying mechanism of the association is also an important priority.


Acknowledgments

The authors appreciate the academic support from the AME Lung Cancer Collaborative Group. The preliminary results of this study were partially published at the American Association for Cancer Research (AACR) Annual Meeting 2022. Abstract ID: 5396. [Cancer Res (2022) 82 (12_Supplement): 5396. https://doi.org/10.1158/1538-7445.AM2022-5396].

Funding: None.


Footnote

Reporting Checklist: The authors have completed the REMARK reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-746/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-746/dss

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-746/coif). YZ, MH, and YB are employed by the company 3D Medicines Inc. The other 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. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). Collection and analysis of data were approved by the Ethics Committee of the First Affiliated Hospital of Guangzhou University of Chinese Medicine (No. K-2022-118). The requirement for informed consent was waived because patients, at the time of treatment, consented for their anonymized medical data to be analyzed and published for research purposes.

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. Mielgo-Rubio X, Uribelarrea EA, Cortés LQ, et al. Immunotherapy in non-small cell lung cancer: Update and new insights. J Clin Transl Res 2021;7:1-21. [PubMed]
  2. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129-39. [Crossref] [PubMed]
  3. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:829-38. [Crossref] [PubMed]
  4. Lim SM, Kim HR, Lee JS, et al. Open-Label, Multicenter, Phase II Study of Ceritinib in Patients With Non-Small-Cell Lung Cancer Harboring ROS1 Rearrangement. J Clin Oncol 2017;35:2613-8. [Crossref] [PubMed]
  5. Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020;382:41-50. [Crossref] [PubMed]
  6. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013;342:1432-3. [Crossref] [PubMed]
  7. Herbst RS, Giaccone G, de Marinis F, et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N Engl J Med 2020;383:1328-39. [Crossref] [PubMed]
  8. Sezer A, Kilickap S, Gümüş M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021;397:592-604. [Crossref] [PubMed]
  9. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019;393:1819-30. [Crossref] [PubMed]
  10. Shields MD, Marin-Acevedo JA, Pellini B. Immunotherapy for Advanced Non-Small Cell Lung Cancer: A Decade of Progress. Am Soc Clin Oncol Educ Book 2021;41:1-23. [Crossref] [PubMed]
  11. Gadgeel S, Rodríguez-Abreu D, Speranza G, et al. Updated Analysis From KEYNOTE-189: Pembrolizumab or Placebo Plus Pemetrexed and Platinum for Previously Untreated Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J Clin Oncol 2020;38:1505-17. [Crossref] [PubMed]
  12. Arbour KC, Riely GJ. Systemic Therapy for Locally Advanced and Metastatic Non-Small Cell Lung Cancer: A Review. JAMA 2019;322:764-74. [Crossref] [PubMed]
  13. Xiong Y, Huang BY, Yin JY. Pharmacogenomics of platinum-based chemotherapy in non-small cell lung cancer: focusing on DNA repair systems. Med Oncol 2017;34:48. [Crossref] [PubMed]
  14. Shukuya T, Carbone DP. Predictive Markers for the Efficacy of Anti-PD-1/PD-L1 Antibodies in Lung Cancer. J Thorac Oncol 2016;11:976-88. [Crossref] [PubMed]
  15. Hsu PC, Jablons DM, Yang CT, et al. Epidermal Growth Factor Receptor (EGFR) Pathway, Yes-Associated Protein (YAP) and the Regulation of Programmed Death-Ligand 1 (PD-L1) in Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2019;20:3821. [Crossref] [PubMed]
  16. Schiller JH, Harrington D, Belani CP, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92-8. [Crossref] [PubMed]
  17. Saijo N, Takeuchi M, Kunitoh H. Reasons for response differences seen in the V15-32, INTEREST and IPASS trials. Nat Rev Clin Oncol 2009;6:287-94. [Crossref] [PubMed]
  18. Olaussen KA, Mountzios G, Soria JC. ERCC1 as a risk stratifier in platinum-based chemotherapy for nonsmall-cell lung cancer. Curr Opin Pulm Med 2007;13:284-9. [Crossref] [PubMed]
  19. Wei Q, Frazier ML, Levin B. DNA repair: a double-edged sword. J Natl Cancer Inst 2000;92:440-1. [Crossref] [PubMed]
  20. Mouw KW, Goldberg MS, Konstantinopoulos PA, et al. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov 2017;7:675-93. [Crossref] [PubMed]
  21. Strickland KC, Howitt BE, Shukla SA, et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016;7:13587-98. [Crossref] [PubMed]
  22. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8. [Crossref] [PubMed]
  23. Chae YK, Davis AA, Raparia K, et al. Association of tumor mutational burden with DNA repair mutations and response to anti–PD-1/PD-L1 therapy in non–small-cell lung cancer. Clin Lung Cancer 2019;20:88-96.e6. [Crossref] [PubMed]
  24. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol 2015;15:760-70. [Crossref] [PubMed]
  25. Härtlova A, Erttmann SF, Raffi FAM, et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 2015;42:332-43. [Crossref] [PubMed]
  26. Ablasser A, Goldeck M, Cavlar T, et al. CGAS produces a 2'-5'-linked cyclic dinucleotide second messenger that activates STING. Nature 2013;498:380-4. [Crossref] [PubMed]
  27. Parkes EE, Walker SM, Taggart LE, et al. Activation of STING-dependent innate immune signaling by s-phase-specific DNA damage in breast cancer. J Natl Cancer Inst 2016;109:djw199. [Crossref] [PubMed]
  28. Scarbrough PM, Weber RP, Iversen ES, et al. A Cross-Cancer Genetic Association Analysis of the DNA Repair and DNA Damage Signaling Pathways for Lung, Ovary, Prostate, Breast, and Colorectal Cancer. Cancer Epidemiol Biomarkers Prev 2016;25:193-200. [Crossref] [PubMed]
  29. Teo MY, Bambury RM, Zabor EC, et al. DNA Damage Response and Repair Gene Alterations Are Associated with Improved Survival in Patients with Platinum-Treated Advanced Urothelial Carcinoma. Clin Cancer Res 2017;23:3610-8. [Crossref] [PubMed]
  30. Hu XC, Zhang J, Xu BH, et al. Cisplatin plus gemcitabine versus paclitaxel plus gemcitabine as first-line therapy for metastatic triple-negative breast cancer (CBCSG006): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 2015;16:436-46. [Crossref] [PubMed]
  31. Pomerantz MM, Spisák S, Jia L, et al. The association between germline BRCA2 variants and sensitivity to platinum-based chemotherapy among men with metastatic prostate cancer. Cancer 2017;123:3532-9. [Crossref] [PubMed]
  32. Choi CM, Yang SC, Jo HJ, et al. Proteins involved in DNA damage response pathways and survival of stage I non-small-cell lung cancer patients. Ann Oncol 2012;23:2088-93. [Crossref] [PubMed]
  33. Li Z, Qing Y, Guan W, et al. Predictive value of APE1, BRCA1, ERCC1 and TUBB3 expression in patients with advanced non-small cell lung cancer (NSCLC) receiving first-line platinum-paclitaxel chemotherapy. Cancer Chemother Pharmacol 2014;74:777-86. [Crossref] [PubMed]
  34. Wang LR, He LJ, Wang Y, et al. Correlation between BRCA1 and TopBP1 protein expression and clinical outcome of non-small cell lung cancer treated with platinum-based chemotherapy. Cancer Chemother Pharmacol 2015;76:163-70. [Crossref] [PubMed]
  35. Corrales L, Glickman LH, McWhirter SM, et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep 2015;11:1018-30. [Crossref] [PubMed]
  36. Ricciuti B, Recondo G, Spurr LF, et al. Impact of DNA Damage Response and Repair (DDR) Gene Mutations on Efficacy of PD-(L)1 Immune Checkpoint Inhibition in Non-Small Cell Lung Cancer. Clin Cancer Res 2020;26:4135-42. [Crossref] [PubMed]
  37. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-47. [Crossref] [PubMed]
  38. Chong CF, Li YC, Wang TL, et al. Stratification of adverse outcomes by preoperative risk factors in coronary artery bypass graft patients: an artificial neural network prediction model. AMIA Annu Symp Proc 2003;160-4. [PubMed]
  39. Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. N Engl J Med 2017;376:2109-21. [Crossref] [PubMed]
  40. Adzhubei IA, Schmidt S, Peshkin L, et al. A method and server for predicting damaging missense mutations. Nat Methods 2010;7:248-9. [Crossref] [PubMed]
  41. Forbes SA, Beare D, Boutselakis H, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 2017;45:D777-83. [Crossref] [PubMed]
  42. Landrum MJ, Lee JM, Benson M, et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 2016;44:D862-8. [Crossref] [PubMed]
  43. Wang C, Yin R, Dai J, et al. Whole-genome sequencing reveals genomic signatures associated with the inflammatory microenvironments in Chinese NSCLC patients. Nat Commun 2018;9:2054. [Crossref] [PubMed]
  44. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:2078-92. [Crossref] [PubMed]
  45. Fan Y, Zhu X, Xu Y, et al. Cell-Cycle and DNA-Damage Response Pathway Is Involved in Leptomeningeal Metastasis of Non-Small Cell Lung Cancer. Clin Cancer Res 2018;24:209-16. [Crossref] [PubMed]
  46. Postel-Vinay S, Soria JC. ERCC1 as Predictor of Platinum Benefit in Non-Small-Cell Lung Cancer. J Clin Oncol 2017;35:384-6. [Crossref] [PubMed]
  47. Pilié PG, Tang C, Mills GB, et al. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol 2019;16:81-104. [Crossref] [PubMed]
  48. Lord CJ, Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017;355:1152-8. [Crossref] [PubMed]
  49. Robson M, Im SA, Senkus E, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 2017;377:523-33. [Crossref] [PubMed]
  50. Teo MY, Seier K, Ostrovnaya I, et al. Alterations in DNA Damage Response and Repair Genes as Potential Marker of Clinical Benefit From PD-1/PD-L1 Blockade in Advanced Urothelial Cancers. J Clin Oncol 2018;36:1685-94. [Crossref] [PubMed]
  51. Zhang Z, Gu Y, Su X, et al. Co-Occurring Alteration of NOTCH and DDR Pathways Serves as Novel Predictor to Efficacious Immunotherapy in NSCLC. Front Oncol 2021;11:659321. [Crossref] [PubMed]
  52. NCCN Clinical Practice Guidelines in Oncology Non-Small Cell Lung Cancer; 2021.
  53. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2019;381:2020-31. [Crossref] [PubMed]
  54. Wang Z, Zhao J, Wang G, et al. Comutations in DNA Damage Response Pathways Serve as Potential Biomarkers for Immune Checkpoint Blockade. Cancer Res 2018;78:6486-96. [Crossref] [PubMed]
  55. Harrison LR, Ottley CJ, Pearson DG, et al. The kinase inhibitor O6-cyclohexylmethylguanine (NU2058) potentiates the cytotoxicity of cisplatin by mechanisms that are independent of its effect upon CDK2. Biochem Pharmacol 2009;77:1586-92. [Crossref] [PubMed]
  56. Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 2007;33:9-23. [Crossref] [PubMed]
  57. Wang W, Figg WD. Secondary BRCA1 and BRCA2 alterations and acquired chemoresistance. Cancer Biol Ther 2008;7:1004-5. [Crossref] [PubMed]
  58. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409-13. [Crossref] [PubMed]
  59. Kitajima S, Ivanova E, Guo S, et al. Suppression of STING Associated with LKB1 Loss in KRAS-Driven Lung Cancer. Cancer Discov 2019;9:34-45. [Crossref] [PubMed]
  60. Brown JS, Sundar R, Lopez J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. Br J Cancer 2018;118:312-24. [Crossref] [PubMed]
  61. Sen T, Rodriguez BL, Chen L, et al. Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer. Cancer Discov 2019;9:646-61. [Crossref] [PubMed]
  62. Klarquist J, Hennies CM, Lehn MA, et al. STING-mediated DNA sensing promotes antitumor and autoimmune responses to dying cells. J Immunol 2014;193:6124-34. [Crossref] [PubMed]
  63. Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol 2016;17:1142-9. [Crossref] [PubMed]
  64. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res 2015;3:436-43. [Crossref] [PubMed]
  65. Knijnenburg TA, Wang L, Zimmermann MT, et al. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 2018;23:239-254.e6. [Crossref] [PubMed]

(English Language Editor: J. Jones)

Cite this article as: Xiao Z, Sun L, Zheng Y, Chen H, Zheng X, Luo J, Gu C, Lin R, Huang M, Bai Y, Chen ZS, Kinslow CJ, Loh J, Lin L. DNA damage repair gene mutations predict the efficacy of platinum-based chemotherapy and immunotherapy plus platinum-based chemotherapy in advanced non-small cell lung cancer: a retrospective Chinese cohort study. Transl Lung Cancer Res 2022;11(12):2539-2566. doi: 10.21037/tlcr-22-746

Download Citation