International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Progress in perioperative immunotherapy for lung cancer and analysis of therapy (Review)
Surgical treatment has long been the preferred approach for lung cancer. In recent years, postoperative adjuvant immunotherapy following surgical treatment has demonstrated good efficacy. Building on this foundation, the introduction of neoadjuvant immunotherapy prior to surgery has also reported promising results in a number of clinical trials. The aim of the present review was to summarize recent research findings associated with perioperative immunotherapy for lung cancer, including key outcomes from both neoadjuvant and adjuvant therapies. The discussion of the results of the present review primarily addressed efficacy, safety, survival data and programmed death‑ligand 1 subgroup analyses. Furthermore, the advantages and limitations of different treatment modalities, including monotherapies and combination therapies, were discussed. Furthermore, the present review incorporated discussions on the applicability of biomarkers in perioperative immunotherapy, the role of immunotherapy combined with other treatments during the perioperative period, mechanisms of resistance and potential solutions to these challenges. In conclusion, the present review offers a systematic examination of the theoretical foundations of perioperative immunotherapy for lung cancer, simultaneously presenting key clinical evidence (including neoadjuvant therapy, adjuvant therapy and ‘sandwich’ combination regimens), diverse combination strategies (chemotherapy and radiotherapy), the current status of biomarker applications and challenges in underlying drug resistance mechanisms for perioperative immunotherapy. It proposes a dynamic monitoring model and clinical decision pathway for omission of adjuvant immunotherapy in non‑small cell lung cancer. Lastly, based upon the latest evidence‑based medical research, the present review aims to provide clinicians and researchers with comprehensive decision‑making references and scientific guidance.
Lung cancer remains the leading cause of cancer-associated mortalities globally, with 2.48 million novel cases and 1.81 million mortalities, recorded in 2022. A model projection released by the International Agency for Research on Cancer under the World Health Organization indicates that new cases of lung cancer will increase by 41% by the year 2050, primarily driven by population aging in China and India. Notably, the nationwide low-dose computed tomography (CT) screening initiative in China has increased the number of cases of early-stage diagnosis by ~12% since 2023 (1–3).
Epidemiological data from 19 countries has revealed that non-small cell lung cancer (NSCLC) accounts for 90.3% of all cases of lung cancer (4). Although surgical resection remains the main curative approach for early-stage and resectable locally advanced NSCLC, postoperative 5-year survival rates remain suboptimal: These have been reported to be 68% for stage IB and 36% for stage IIIA disease, respectively (5). Although traditional adjuvant chemotherapy has been reported to modestly reduce the risk of recurrence, its clinical benefits appear to have reached a plateau, since only a 5% improvement in 5-year survival rates has been recorded (6), with additional limitations including chemotherapy-associated toxicity and the absence of validated predictive biomarkers. Notably, 30–55% of patients experience locoregional recurrence or distant metastasis postoperatively (7), underscoring the key need for effective eradication of minimal residual disease (MRD) and the activation of systemic immune surveillance to improve outcomes.
The advent of immune checkpoint inhibitors (ICIs) targeting programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) has revolutionized treatment paradigms for advanced NSCLC (8). Key phase III trials, including KEYNOTE-024 and CheckMate 227 (9,10), have demonstrated notable overall survival (OS) benefits with ICI monotherapy or combination regimens in metastatic settings. These breakthroughs have prompted an investigation of immunotherapy in different perioperative settings, aiming to remodel the tumor immune microenvironment through neoadjuvant or adjuvant approaches to achieve synergistic locoregional-systemic control.
Early exploratory studies, such as the LCMC3 and CheckMate 159 studies (11,12), were able to establish the feasibility of neoadjuvant immunotherapy. The landmark CheckMate 816 trial first demonstrated in a phase III setting that the use of the checkpoint inhibitor nivolumab plus chemotherapy led to a marked improvement in both the pathological complete response (pCR) rates and event-free survival (EFS) rates compared with chemotherapy alone in resectable NSCLC, marking the transition of perioperative immunotherapy from the conceptual stage to clinical practice (13).
Subsequent trials, including the KEYNOTE-671 (14) and AEGEAN (15) trials, further advanced combination strategies, achieving pCR rates >30% and demonstrating durable survival benefits with postoperative adjuvant immunotherapy. Similar progress in adjuvant immunotherapy has emerged from the IMpower010 trial, where treatment with atezolizumab led to an improved 3-year disease-free survival (DFS) rate (increased to 60%) compared with chemotherapy in PD-L1+ (≥1%) patients with stage II–IIIA cancer (16), whereas the KEYNOTE-091 trial established a DFS benefit for pembrolizumab in PD-L1-unselected cases. These findings have been incorporated into major guidelines, including those of the National Comprehensive Cancer Network (NCCN) and the International Association for the Study of Lung Cancer (IASLC) (17–19).
Nevertheless, several challenges persist in this rapidly evolving field. First, the selection of biomarkers remains contentious: The CheckMate 816 trial revealed marked pCR rate disparities comparing between the PD-L1 ≥1% and PD-L1 <1% subgroups (13), whereas the KEYNOTE-091 trial revealed PD-L1-independent DFS benefits. Secondly, safety optimization of combination strategies warrants urgent attention, as exemplified by the 32% grade 3–4 adverse event (AE) rate with dual immunotherapy that was observed in the CheckMate 77T trial (20). Furthermore, the limited efficacy of ICIs noted in epidermal growth factor receptor (EGFR)/anaplastic lymphoma kinase (ALK)-altered NSCLC needs to be followed up with mechanistic studies (21). Lastly, long-term follow-up data are required to fully characterize the impact of immune-associated AEs (irAEs) on perioperative outcomes (22).
Currently, the paradigm of perioperative management is expanding toward integrated systemic approaches that target not only tumor cells, but also the microenvironment of the host. Increasing evidence has highlighted the pivotal role of systemic inflammation and metabolic dysregulation in modulating treatment efficacy; for example, an elevated neutrophil-to-eosinophil ratio was reported to be associated with impaired OS [hazard ratio (HR)=1.74] and progression-free survival (PFS; HR=1.53) rates across malignancies, reflecting pro-tumorigenic inflammation that may undermine immunotherapeutic responses (23). In addition, metabolic syndrome, which is characterized by chronic inflammation and insulin resistance, has been reported to increase cancer risk via adipokine dysregulation and oxidative stress, thereby suggesting that metabolic interventions may potentially act synergistically with ICIs (24).
Concurrently, novel biomarkers are refining patient stratification. The cachexia index, which integrates skeletal muscle mass, albumin and neutrophil-associated lymphocytes, predicts survival rates (OS, HR=2.03) independently of tumor stage, thereby offering a multidimensional assessment of the nutritional-inflammation status (25). Despite these emerging tools; however, PD-L1, as a biomarker, retains its clinical relevance in cases of advanced NSCLC: A high expression (≥50%) of PD-L1 remains predictive of notably increased ICI responsiveness, warranting a further exploration of its dynamic changes in perioperative settings (26). These insights align with other therapeutic advances that have recently been made; for example, meta-analyses have confirmed that adjuvant PD-1/PD-L1 inhibitors reduce the recurrence-free survival (RFS) risk across solid tumors (HR=0.72), thereby supporting their broader application beyond PD-L1 selection (27).
Future research efforts should prioritize precision medicine through the application of multi-omics approaches [for example, single-cell sequencing and spatial transcriptomics (28)] to decipher dynamic tumor-immune microenvironment interactions, to develop circulating tumor DNA (ctDNA)-based MRD monitoring systems and to explore novel therapeutic combinations [for example, immunotherapy with targeted agents (29) or epigenetic modulators (30)] to overcome drug resistance. Real-world evidence will be key to the validation of treatment paradigms in special populations. Subsequently, the present review will systematically examine recent advances that have been made in perioperative immunotherapy through four dimensions, namely clinical applications, combination strategies, biomarker development and future challenges, with the aim of providing information for evidence-based clinical decision-making in the future.
The present narrative review has compiled evidence from pivotal clinical trials published between January 1, 2007 and August 31, 2025, using the search terms ‘perioperative immunotherapy’, ‘neoadjuvant/adjuvant’, ‘lung cancer’ and ‘clinical trial’. Key sources included the PubMed (https://pubmed.ncbi.nlm.nih.gov/) and Web of Science (https://www.webofscience.com) databases, proceedings from major oncology conferences (for example, those organized by American Society of Clinical Oncology (ASCO) (31–33) and European Society for Medical Oncology (ESMO) (34) and IASLC (35,36) and American Association of Cancer Research (AACR) (37) and the Clinical Trials website (clinicaltrials.gov) (for ongoing trials). Phase III randomized controlled trials (for example, CheckMate816 and KEYNOTE-671) were prioritized, along with practice-changing phase II studies that have informed recent guideline updates (for example, the updates made to version 7.2025; NCCN (18) and Neoadjuvant and Adjuvant Treatments for Early-Stage Resectable NSCLC Consensus Recommendations From IASLC (19). In addition, emphasis was placed on perioperative outcomes (regarding parameters such as the pCR and the EFS/OS rate), biomarker correlates (PD-L1 and ctDNA) and the safety profiles of immunotherapy combinations.
The cancer-immunity cycle, a fundamental biological process governing immune-mediated tumor recognition and eradication, comprises seven sequential steps, namely tumor antigen release, antigen presentation, T-cell activation, T-cell trafficking to tumor sites, tumor-infiltrating T-cell recognition of cancer antigens, tumor cell killing and the establishment of immunological memory. ICIs primarily function by counteracting immunosuppressive signals within the tumor microenvironment (TME), thereby reactivating antitumor immune responses (Fig. 1) (38–44). However, the seven steps in the cancer-immunity cycle of lung cancer tumors are notably specific and different perioperative immunotherapy approaches work through a range of different mechanisms (Table I) (45–50).
Table I.Lung cancer-specific mechanism in the cancer-immunity cycle and perioperative immunotherapy measure. |
The exploration of ICIs in neoadjuvant therapy began with early single-agent clinical studies, aimed at evaluating their potential to induce pathological remission and assessing their safety. Although neoadjuvant immunotherapy combined with chemotherapy has become the predominant strategy, research on neoadjuvant monotherapy immunotherapy continues to provide key insights into the biological effects of immunotherapy and patient selection.
In the CheckMate 159 trial (12), the major pathological response (MPR) rate of nivolumab monotherapy was reported to reach 45% and patients with a high tumor mutational burden (TMB) exhibited notable pathological remission. Furthermore, the 5-year RFS risk for PD-L1+ patients was reduced by 64% (HR=0.36), whereas the MPR rate for PD-L1− patients was 30%, demonstrating that PD-L1− patients could also derive certain benefit from the therapy. Furthermore, the LCMC3 study (11) further validated the feasibility of neoadjuvant immunotherapy monotherapy. The MPR rate of atezolizumab monotherapy was reported to be 20%, although this increased significantly to 33% in the PD-L1 ≥50% subgroup compared with only 11% in the tumor proportion score (TPS) <1% group (P=0.01). The MPR rate in the TMB ≥16 mutant/Mb subgroup was also 33%, whereas it was only 13% in the low TMB group (P=0.12), highlighting the synergistic screening potential of dual markers. Furthermore, compared with traditional neoadjuvant chemotherapy studies that had an MPR rate of 19% and a treatment-related AE (TRAE) rate of 57%, (51), the MPR rate of neoadjuvant monotherapy immunotherapy increased to 20–45%, with markedly reduced toxicity. The incidence of grade 3 TRAEs was 11–23%, which was notably lower than the 60% incidence of TRAEs observed in neoadjuvant chemotherapy. Notably, the surgical delay rate in the LCMC3 study was extremely low. Similarly, in the NEOSTAR monotherapy group (52), the MPR rate was 22%, while the pCR rate was 9%, both markedly higher compared with those of patients receiving traditional chemotherapy. However, there remained a gap in the efficacy of monotherapy compared with the dual immunotherapy combination: For example, the MPR rate of the nivolumab plus ipilimumab combination increased to 38%, whereas that of the monotherapy group was only 22% and the dual immunotherapy combination also led to the induction of higher levels of effector T cell infiltration, with a 50% increase in CD8+ T cells compared with neoadjuvant immunotherapy monotherapy (P=0.033) (34,52–54).
In general, the pCR of the neoadjuvant monotherapy immunotherapy was reported to be low (generally of the order of <10-15%). Furthermore, the pCR was notably lower compared with that of immunotherapy combined with chemotherapy; for example, the MPR in the CheckMate 816 trial was reported to be 24%. It should be noted that the majority of the single-agent studies had small sample sizes and limited representativeness (Table II).
At present, a combination of ICIs and chemotherapy has become the standard regimen for the neoadjuvant treatment of resectable NSCLC (55). This strategy works by enhancing tumor antigen release via chemotherapeutic-induced immunogenic cell death (ICD) and synergistically activating systemic antitumor immune responses with ICIs, thereby causing marked improvements in pathological remission rates and long-term survival.
Global studies have demonstrated that the pCR rate of nivolumab in combined chemotherapy was 24%, markedly higher compared with the 2.2% reported for the chemotherapy control group and the MPR rate was 34%, also much higher compared with the MPR rate of 12% recorded for the chemotherapy control group. Of note, the pCR rate of Chinese patients was as high as 25% (13,56). Furthermore, in the LungMate 002 study (50), the pCR rate for the neoadjuvant toripalimab plus chemotherapy subgroup was reported to be as high as 27.8% and the MPR rate was as high as 55.6%. Patients with high baseline expression of chitinase 3-like protein 1 (CHI3L1) [according to the immunohistochemical (IHC) score] exhibited significantly improved pCR and survival outcomes (for OS, HR=0.17 and P=0.017), demonstrating that CHI3L1 can serve as an independent predictive marker. By contrast, the pCR rate for neoadjuvant immunotherapy alone was only 10–15%, whereas the pCR improvement following neoadjuvant immunotherapy combined with chemotherapy was notable (the combination therapy group achieved 27.8% pCR in LungMate 002, while the pCR rate in the combination therapy group was 24% in CheckMate 816), highlighting the enhanced efficacy of the combined regimen.
Regarding surgical feasibility, in the CheckMate 816 study, the surgery rate for the immunotherapy combined with chemotherapy group was reported to be 83.2% (compared with 75.4% for the chemotherapy alone group) and more minimally invasive surgeries were performed. Furthermore, in the LungMate 002 study, 65.8% of the 76% of the patients who were potentially resectable in the neoadjuvant immunotherapy combined with chemotherapy group were converted into operable patients, achieving a total R0 resection rate of 100% (where ‘R0 resection’ refers to a surgical outcome where all the cancerous tissue is removed). Additionally, the administration of neoadjuvant immunotherapy combined with chemotherapy achieved a lymph node downstaging rate of 63.9%, suggesting that this regimen led to a marked improvement in surgical feasibility via the reduction of tumor size and downstaging.
In terms of safety, the incidence of grade 3–4 TRAEs in the neoadjuvant immunotherapy combined with chemotherapy group in the CheckMate 816 study was reported to be 33.5%, a percentage that was similar to the reported percentage of 36.9% in the chemotherapy alone group, and neoadjuvant immunotherapy combined with chemotherapy did not lead to increases in the rates of surgical complications or delays. In the LungMate 002 study, the incidence of grade 3–4 TRAEs in neoadjuvant immunotherapy combined with chemotherapy was reported to be 34%. Overall, compared with monotherapy immunotherapy, the chemotoxicity of combined chemotherapy was reported to be slightly higher, although its safety may have been well controlled through standardized management and this toxicity did not markedly affect surgical safety. By contrast, monotherapy immunotherapy may lead to a higher risk of tumor progression due to insufficient efficacy. Subgroup analysis revealed that the pCR rate (partial response to curative therapy) for patients with PD-L1 levels ≥1% in the CheckMate 816 trial was 32.6%, markedly higher compared with the pCR rate of 16.7% observed in the subgroup with PD-L1 levels <1%. In summary, the administration of immunotherapy combined with chemotherapy causes the release of tumor antigens, thereby enhancing immune responses and compensating for the insufficient immune response rate in neoadjuvant monotherapy, which provides a focus in the field of neoadjuvant immunotherapy with more potential benefits than drawbacks (Table II) (11–13,34,57–63).
The combined use of RT and ICIs is based on the principle that RT induces ICD in tumor cells, leading to the release of damage-associated molecular patterns, which, in turn, enhance the antigen-presenting ability of dendritic cells (DCs). ICIs subsequently further alleviate systemic immunosuppression, creating an ‘in situ vaccine’ effect (64).
Key studies have validated the clinical potential of this strategy. In the SQUAT trial (65), the MPR rate of the neoadjuvant durvalumab combined with concurrent chemoradiotherapy (50 Gy) was reported to be 63%, which was notably higher compared with the MPR rate of 36.8% reported in the CheckMate 816 trial for neoadjuvant immunotherapy plus chemotherapy. Furthermore, the pCR rate in the SQUAT trial was 23%, similar to the 24% pCR rate noted in the CheckMate 816 trial, indicating that neoadjuvant immunotherapy combined with chemotherapy led to a notable enhancement in the extent of pathological remission (65). In terms of long-term survival, compared with traditional RT, the objective response rate (ORR) for the KEYNOTE-799 regimen with concurrent chemoradiation therapy (cCRT) plus pembrolizumab was 70.5–70.6%, whereas the ORR for the NICOLAS regimen with cCRT plus nivolumab was 73.4%. Both regimens were noted to have higher ORRs compared with the 35–55% range observed in traditional chemoradiotherapy (66,67).
In terms of safety, the SQUAT trial reported a 48% rate of grade 3–4 AEs (including only 1 case of fatality that was associated with the treatment), which was higher compared with that of chemotherapy alone or monotherapy immunotherapy. However, the toxicity profile was reported to be similar to that of historical Neoadjuvant Chemoradiotherapy data (68). Furthermore, the most notable AE arising from neoadjuvant immunotherapy combined with RT was shown to be radiation pneumonitis. The incidence of grade 3 or higher pneumonia in the KEYNOTE-799 and NICOLAS combination regimens was 6.9% and up to 10%, respectively. By contrast, the incidence of grade 3 or higher radiation pneumonitis in traditional chemoradiotherapy is typically reported to be between 7 and 20% (69). This suggests that the addition of RT did not exceed the safety threshold. In terms of surgical feasibility, all patients in the SQUAT trial completed their surgeries on schedule, with no reports of surgery delays due to RT.
Regarding the expression of PD-L1, in the KEYNOTE-799 study, the ORR for patients with PD-L1 TPS <1% in cohorts A and B was reported to be 66.7 and 71.4%, respectively, whereas for those patients with PD-L1 ≥1% in cohorts A and B, the ORR was 75.8 and 72.5%, respectively. This demonstrates that there is minimal difference in efficacy among patients with different expression levels of PD-L1. Although the SQUAT trial did not subdivide the PD-L1 groups, the overall MPR rate was still as high as 63%, indicating its broad-spectrum efficacy. However, although the SQUAT trial demonstrated a high MPR rate of 63% for neoadjuvant chemoradiotherapy combined with immunotherapy, the 2-year PFS rate was only 43% and no notable improvements were observed in terms of patient survival. This may suggest that, although RT enhances local immune activation, it may not effectively eliminate MRD. Additionally, the SQUAT trial did not report distant recurrence rates, suggesting that RT might either induce T-cell exhaustion or upregulate PD-L1, thereby weakening the systemic effects of durvalumab and potentially affecting the long-term prognosis of patients.
The results from IMpower010 (16), a randomized, multicenter, open-label phase 3 trial, demonstrated that postoperative adjuvant chemotherapy followed by sequential atezolizumab treatment led to a notable reduction in the recurrence risk in the PD-L1 ≥50% subgroup (DFS, HR=0.48; OS, HR=0.47). Compared with the Best Supportive Care (BSC) group, which was reported to have a 5-year DFS rate of only 67.5%, the 5-year DFS rate for the adjuvant atezolizumab treatment group increased to 84.8% and distant metastasis was well controlled. The distant metastasis recurrence rate in the adjuvant atezolizumab treatment group was reported to be 6.8% compared with 12.5% in the BSC group, with particularly good control exerted over brain metastasis recurrence. Furthermore, in another study, KEYNOTE-091 (17), treatment postoperatively with the adjuvant pembrolizumab demonstrated DFS benefits in the overall population (HR=0.76), although this did not reach significance in the PD-L1 ≥50% subgroup (HR=0.82).
In terms of safety, the incidence of grade 3–4 irAEs in the IMpower010 study was only 11%, compared with an incidence of 25% for cisplatin-associated grade 3–4 nausea/vomiting when traditional adjuvant therapy was administered. This suggests that adjuvant immunotherapy is both safer and more tolerable compared with traditional adjuvant chemotherapy. In the KEYNOTE-091 trial, although the incidence of grade 3–4 irAEs was higher (at 34%) among patients receiving postoperative adjuvant immunotherapy, only 4 cases were reported to be associated with myocarditis or pneumonia. This indicated that fatal irAEs are rare and that the majority of irAEs are manageable, rendering them easier to manage compared with the long-term toxicities associated with chemotherapy, such as kidney damage (70) and hearing loss (71). Notably, in both the IMpower010 and the KEYNOTE-091 trials, administering sequential immunotherapy following chemotherapy did not lead to any notable increase in cumulative toxicity, supporting the synergistic model of chemotherapy clearing the immunosuppressive microenvironment combined with immunomodulation (16,17). In addition to the aforementioned postoperative adjuvant immunotherapy, clinical trials of postoperative adjuvant immunotherapy in combination with other regimens, such as INTerpath-002 and SKYSCRAPER-15, have gradually revealed their application prospects (Table III) (16,17,31,32,35–37,72–75).
The ‘sandwich model’ of perioperative immunotherapy for lung cancer comprises a comprehensive strategy that includes preoperative neoadjuvant immunotherapy combined with chemotherapy, surgical resection and postoperative adjuvant immunotherapy to consolidate treatment. This approach aims to activate the immune system preoperatively to shrink tumors and to reduce the stage of cancer, to achieve radical resection during surgery and to continuously eliminate MRD postoperatively.
In the NEOTORCH trial (a randomized, double-blind, placebo-controlled phase 3 trial), the triple therapy group achieved a pCR rate of 24.8%, whereas in the RATIONALE-315 trial (another randomized, double-blind, placebo-controlled phase 3 trial), this rate increased to 56% (76,77). By contrast, in the LCMC3 study, the pCR rate for patients receiving only immunotherapy as neoadjuvant treatment was only 6% (11), and in the CheckMate 816 trial (13), the pCR rate for patients receiving combined immunotherapy and chemotherapy as neoadjuvant treatment was only 24%. Furthermore, in the KEYNOTE-671 study (14), the MPR rate for patients treated with the triple therapy model reached 63.8%, in the RATIONALE-315 trial it was 56% and in the NEOTORCH trial it was 48.5%; by contrast, in the CheckMate 816 study, the MPR rate for patients receiving combined immunotherapy and chemotherapy as neoadjuvant treatment was only 24%, and in the NEOSTAR study, the MPR rate for the single-agent neoadjuvant immunotherapy group was only 22%. This clearly demonstrates that, in terms of pathological remission, the triple therapy model is markedly more effective compared with monotherapy neoadjuvant treatment (13,52,76,77).
Additionally, from the perspective of surgical feasibility, the surgical resection rate for patients treated with the triple therapy model in the KEYNOTE-671 trial was as high as 93%; in the CheckMate 77T study, it was 89%; in the RATIONALE-315 study, it was 91%; whereas in the CheckMate 816 trial, the surgical resection rate for patients receiving combined immunotherapy and chemotherapy as neoadjuvant treatment was only 83.2%. According to the subgroup analysis of CheckMate 816 trial, similarly to the results in the single neoadjuvant setting, the sandwich model also demonstrated a more notable benefit for patients with PD-L1 levels of ≥1%. The EFS HRs were determined to be 0.52 and 0.58 in the CheckMate 77T and KEYNOTE-671 trials, respectively. However, in the subgroup with PD-L1 levels <1%, the HRs increased to 0.73 and 0.65, suggesting limited benefits for patients with PD-L1 levels <1%. Nevertheless, the RATIONALE-315 study demonstrated that patients with PD-L1 levels <1% still obtained certain benefit (EFS, HR=0.57). This benefit was primarily due to the unique mechanism of action of tislelizumab, whereby antibody-dependent cell-mediated phagocytosis is reduced, thereby overcoming the limitations of low PD-L1 expression and providing treatment opportunities for these patients.
Notably, tislelizumab has been underutilized in perioperative immunotherapy studies for lung cancer, with potential for enhanced research on this ICI in later stages. Furthermore, compared with the IMpower010 study, in which a simple adjuvant therapy was administered, the survival benefits of the sandwich model were more pronounced. In the NEOTORCH study, the EFS HR for patients treated with the sandwich model was 0.40; in the KEYNOTE-671 study, it was 0.59; and in the RATIONALE-315 study, it was 0.56. By contrast, in studies of adjuvant immunotherapy alone, the DFS HR for all patients receiving postoperative adjuvant immunotherapy in the IMpower010 study was 0.79 and in the KEYNOTE-091 study, the overall DFS HR was 0.76. Although the primary endpoints of the sandwich model study and the adjuvant postoperative treatment study differed, the EFS parameter includes a broader range of events, such as local recurrence, distant metastasis, second primary cancer and treatment-associated mortality, rendering it more stringent compared with DFS, which only includes recurrence/mortality. However, the sandwich model was still reported to have an improved HR, demonstrating its ability to comprehensively control tumor progression.
It should be noted that, although the sandwich model exhibited a notably increased efficacy compared with simple adjuvant and neoadjuvant immunotherapy, it may face greater challenges in terms of safety management. In the tislelizumab group of the RATIONALE-315 trial, the incidence of irAEs was reported to be 40%, a percentage that was markedly higher than the incidence rate of 18% for irAEs in the chemotherapy group. Furthermore, the incidence of grade 3 or higher neutropenia in the tislelizumab group was as high as 61% and these patients required close monitoring and management. Additionally, administering long-term adjuvant immunotherapy may increase the burden on patients. For example, in the NEOTORCH study, the sandwich model treatment regimen included treatment with 13 cycles of toripalimab. By contrast, the treatment may be prematurely discontinued for certain patients due to toxicity. For example, in the CheckMate 77T study, 35% of the patients did not complete the course of postoperative adjuvant therapy due to intolerance to treatment-associated toxicity (14,20,76,77).
In conclusion, perioperative immunotherapy, which involves tumor shrinkage prior to surgery and continuous immunosuppression following surgery, markedly outperforms preoperative or postoperative adjuvant therapy in terms of pathological remission and survival benefits. It is suitable for most high-risk patients who are able to tolerate toxicity, particularly in the cases of borderline resectable patients, to improve surgical outcomes. For patients with PD-L1 levels ≥1%, it is recommended to prioritize dual immunotherapy or combination chemotherapy. These two treatment regimens were reported to achieve marked pathological remission in the NEOSTAR and RATIONALE-315 studies. Even for the PD-L1− patients, the sandwich model maintained its superiority compared with postoperative adjuvant therapy; for example, in the CheckMate 77T trial, the EFS HR for patients in the PD-L1 <1 group was only 0.65 (Table IV) (12,15,20,33,76–83).
As discussed previously, employing the sandwich therapy regimen leads to a notable improvement in pathological response rates and survival outcomes, although it also presents greater safety challenges compared with administering monotherapy with neoadjuvant immunotherapy. First, the sandwich therapy regimen is associated with markedly higher rates of TRAEs, particularly irAEs, compared with monotherapy. Furthermore, its extended treatment duration markedly increases the risks of cumulative toxicity, leading to its discontinuation during the adjuvant immunotherapy phase, thereby imposing heavier financial burdens on patients. The current consensus recommends neoadjuvant immunotherapy combined with chemotherapy for all patients with operable stage II–III lung cancer. However, whether postoperative immunotherapy should be upgraded to sandwich therapy remains a subject of ongoing debate (19,55,84). Therefore, the findings of recent studies on perioperative immunotherapy for lung cancer have led to the development of the clinical decision pathway for omission of adjuvant immunotherapy in NSCLC (Fig. 2). The core criteria for pathogenesis-guided adjuvant immunotherapy are pCR with postoperative sustained ctDNA negativity or switching to targeted therapy in driver gene-cases. High-risk patients (namely, those with stage III lung cancer, non-pCR patients and those with high expression of PD-L1) should still complete standardized 1-year adjuvant therapy; however, it must be emphasized that, even in terms of the pCR, patients with squamous cell carcinoma still face a 5-year recurrence risk >30%. Therefore, maintaining full adjuvant therapy is recommended. The current strategy faces challenges due to limitations in pCR assessment (at present, ~8% of micro-metastases are missed) and variations in ctDNA detection sensitivity (currently, the false negative rate is in the range 5–10%). Thus, decision-making regarding exemption requires multidisciplinary team consultation (84–88).
Biological markers perform a key role in perioperative immunotherapy, which is aimed at identifying potential beneficiaries, optimizing treatment decisions and predicting the risk of drug resistance. Currently, the most extensively studied markers are PD-L1 expression, TMB and dynamic monitoring of ctDNAs. However, their clinical application faces numerous challenges. Emerging biomarkers, such as Gene Expression Omnibus (GEO) profiles and tumor-infiltrating lymphocytes (TILs), are beginning to gain prominence.
In perioperative immunotherapy for lung cancer, PD-L1, ctDNAs and TMB each have unique value as biomarkers, although each of them has its limitations. PD-L1 expression is the most widely used biomarker. The CheckMate 816 study demonstrated that patients with PD-L1 levels of ≥1% had a pCR rate of 32.6%. The KEYNOTE-671 study further confirmed that patients with high PD-L1 expression (TPS ≥50%) received more notable survival benefits. Specifically, in patients treated with the sandwich model, those with low PD-L1 expression (TPS <50%) had an EFS HR of 0.61, whereas those with high PD-L1 expression had an EFS HR of only 0.41 (13,14). However, in the NEOTORCH study, no clear association was identified between EFS benefit and PD-L1 expression. Although the EFS HR values for the subgroup with PD-L1 levels ≥50% and the sandwich model treatment subgroup with a PD-L1 level <50% were 0.31 and 0.35 respectively, neither of these subgroups reached the level of statistical significance, suggesting that PD-L1 expression levels may not serve as an independent predictor of efficacy (77). Furthermore, the spatiotemporal heterogeneity of PD-L1 limits its predictive power, with temporal heterogeneity indicating that PD-L1 expression may vary dynamically with treatment. For example, preoperative neoadjuvant therapy may alter the TME, leading to changes such as TILs and the release of inflammatory factors. This can result in the measured postoperative PD-L1 expression levels in specimens being inconsistent with the baseline levels of patients, thereby reducing the predictive value of PD-L1. The spatial heterogeneity is evident in notable differences in PD-L1 expression being reported among different regions within the same tumor or between the primary lesion and metastatic sites. Therefore, biopsy specimens may only reflect local expression status, rather than the overall tumor characteristics (89).
As biomarkers, ctDNAs have performed particularly well with respect to MRD monitoring. The CheckMate 816 subgroup analysis revealed that patients who tested positive for ctDNA post-surgery had an EFS event rate of 72.1% within 12 months, compared with an EFS event rate of only 14.3% for those who tested negative. Furthermore, the exploratory analysis of the KEYNOTE-671 study indicated a positive association between ctDNA clearance and the pCR rate following neoadjuvant immunotherapy. In the ctDNA clearance group where ctDNA was undetectable post-treatment, the pCR rate reached 46.8%, whereas in the non-clearance group, where ctDNA remained detectable post-treatment, the pCR rate was reported to be only 7.7% (11,12). Therefore, the ‘TNMB staging’ model, combining ctDNA and TNM staging, was proposed in the MEDAL study, in which the results reported that the model was capable of improving the accuracy of recurrence prediction by 28% (90).
TMB is a surrogate indicator of tumor neoantigen load. The CheckMate 227 study demonstrated that, among patients with high TMB (≥10 mut/Mb), those treated with nivolumab plus ipilimumab achieved a median PFS of 7.2 months, whereas it was only 5.5 months in those with low TMB (<10 mut/Mb; HR=0.58). This finding suggests that TMB may serve as a potential predictor of short-term efficacy for immune-combination therapy (10). However, several perioperative studies lack a standardized TMB testing standard (10,11). Whole exome sequencing, which covers ~30,000 genes, does provide a high-precision assessment of TMB, although it is costly and complex, making it difficult to implement the method widely. By contrast, TMB may be inferred from lower-cost targeted panel sequencing, which covers limited genomic regions (typically 1–2 Mb), using the application of fitting algorithms. However, different panel designs can lead to biased results; for example, panels with smaller coverage areas may underestimate TMB values. Furthermore, the TMB thresholds have been reported to vary across different detection platforms; for example, the FoundationOne®CDx platform (Foundation Medicine) detects tissue TMB (tTMB) with a preset threshold of 10, 13 or 16 mut/Mb for tTMB, whereas a preset threshold of 20 mut/Mb is used for blood TMB (bTMB). Although all methods are able to distinguish between high and low TMB levels, inconsistent thresholds have been reported to result in varying clinical benefits. Currently, whole-exome sequencing (WES) and large-panel sequencing are the most commonly used methods for measuring TMB. However, a bridging study involving patients with NSCLC found that 199 missense mutations detected by WES corresponded to a TMB of 10 mut/Mb measured using the FoundationOne CDx (F1 CDx) panel. Although the overall consistency between the two methods reached 84, ~16% of patients may still be misclassified as having low or high TMB due to platform differences. Furthermore, in Cohort C of the BFAST trial, atezolizumab was compared with chemotherapy as first-line therapy for patients with unresectable, advanced-stage NSCLC characterized NSCLC characterized by a bTMB cutoff of ≥10 mut/Mb. Interestingly, in an exploratory analysis where bTMB was evaluated using the F1L CDx assay with an equivalent cutoff of bTMB ≥13.60 mut/Mb, atezolizumab significantly improved PFS compared with chemotherapy. These examples indicate that even within the same population, differences in threshold settings across platforms may result in some patients being unable to be accurately matched with the most suitable treatment regimen (91,92).
Gene expression profiling (GEP) analysis comprises an exploration of immune-associated gene expression patterns in the TME using high-throughput sequencing technology, including the analysis of biological markers such as the interferon-γ (IFN-γ) signaling pathway, T cell activation-associated genes [including CD8A and granzyme B (GZMB)] and immunosuppressive molecules [such as TGF-β and indoleamine 2,3-dioxygenase 1 (IDO1)] (93). In the LungMate 002 study, a receiver operating characteristic curve was constructed by comparing the IHC scores of baseline CHI3L1 with the post-treatment pathological remission status. The area under the curve was calculated to be 0.732, indicating that the IHC score of CHI3L1 in baseline tumor samples can predict the treatment response. Patients with high CHI3L1 expression were reported to have an OS HR of 0.17 and a PFS HR of 0.29, suggesting that patients with high CHI3L1 expression had improved survival outcomes. Even in patients with PD-L1− expression, those with high CHI3L1 expression still derived a notable benefit from perioperative immunotherapy, suggesting that CHI3L1 was able to effectively address the prediction ‘blind spot’ of PD-L1. This is partly because CHI3L1 may influence matrix remodeling by regulating immune-suppressive cells in the TME, thereby enhancing the synergistic effect of chemotherapy combined with immunotherapy.
Furthermore, PD-L1 only reflects the expression of immune checkpoints on the surface of tumor cells and does not fully capture the overall immune activity in the TME. Additionally, in the LungMate 002 study, a high expression of human leukocyte antigen (HLA)-DR was observed in the response group. HLA-DR, a class II major histocompatibility complex (MHC) class II cell surface receptor, performs a key role in antigen presentation and in the activation of CD4+ T cells. In this study, a regimen of toripalimab combined with chemotherapy caused the release of tumor antigens by administering chemotherapy, which, when combined with PD-1 inhibitors to block immune checkpoints, led to a synergistic enhancement of HLA-DR-mediated antigen presentation, forming an immune positive feedback loop. Furthermore, it was reported that CHI3L1 is positively associated with HLA-DR expression, suggesting that CHI3L1 may indirectly promote HLA-DR-mediated antigen presentation by regulating tumor matrix remodeling (57). In addition, in the NEOSTAR dual immunotherapy group, a notable increase in GZMB upregulation was observed. This upregulation of GZMB was reported to be associated with the expansion of effector memory T cells (TEM) and tissue-resident memory T cells (TRM) within tumors, suggesting that the upregulation of GZMB may enhance T-cell function. Furthermore, patients with high GZMB expression had a median viable tumor percentage of only ~9% compared with the percentage of 50% observed in the single-agent immunotherapy group. This suggested that GZMB-mediated T-cell toxicity is a key driver of pathological remission. Additionally, in the dual immunotherapy group, the mRNA levels of C-X-C motif chemokine ligand (CXCL) 9 and CXCL10 were markedly elevated. In this study, IHC analysis was also performed, which confirmed that the protein expression levels of CXCL9 and CXCL10 in the tumor tissues were higher compared with those of the dual immunotherapy group, particularly in patients with pathological remission. This may have been due to ipilimumab blocking the inhibitory signals of regulatory T cells (Tregs), thereby enhancing the antigen-presenting capability of the DCs and promoting the secretion of CXCL9/10 (67).
TILs are lymphocytes that migrate from the blood into tumor tissues, where they serve as a key component of the TME. These cells have a dual regulatory role in immune response through their recognition of tumor antigens, which is key to the development, progression and treatment of tumors. TILs primarily include CD8+ T cells, CD4+ T cells, Tregs and natural killer (NK) cells. In the CheckMate 159 study, it was observed that, following immunotherapy, the number of neoantigen-specific T cells in both tumor tissues and peripheral blood were markedly increased. These increases were particularly pronounced in patients who had achieved pCR, suggesting that neoantigen-specific T cells may directly contribute to tumor clearance. Additionally, in the NEOSTAR study, the combined immunotherapy group exhibited a marked increase in the density of CD8+ T cells infiltrating the tumor compared with the monotherapy immunotherapy group. The proportions of TRM and TEM were also increased, whereas the levels of Tregs and myeloid-derived suppressor cells (MDSCs), which are immunosuppressive cells, were decreased. Furthermore, in the LCMC3 study, it was reported that the density of Tregs and MDSCs in the TME was negatively associated with the pathological response; specifically, the enrichment of MDSCs was reported to be notably associated with a low MPR rate for atezolizumab. In addition, it was reported that immunoglobulin-like transcript 2 T cells were also negatively associated with the MPR. The LCMC3 study also demonstrated that the proportions of NK group 2 member D and non-T/NK cells, including γδ T cells or congenital lymphocytes, in baseline peripheral blood were positively associated with the MPR (11,12) (Table V) (32,36,75,79).
In addition to the aforementioned GEP analyses and TILs, the NEOSTAR study also demonstrated that the abundance of Ruminococcus and Akkermansia in the gut microbiota is notably associated with the MPR of dual immunotherapy. This finding suggests that specific bacterial populations may enhance antitumor effects by modulating systemic immune responses (45), indicating that these specific bacterial populations may have notable potential as biomarkers for perioperative immunotherapy.
Current evidence suggests that integrated biomarker models are able to enhance prediction accuracy (94). Therefore, our research group developed a dynamic monitoring model for perioperative immunotherapy in lung cancer that works through integrating mainstream non-treatment biomarkers.
First, ctDNA clearance rates were assessed in patients with advanced unresectable lung cancer after one cycle of neoadjuvant immunotherapy. Those achieving clearance continued with the original treatment regimen, whereas those without clearance either received an additional cycle of neoadjuvant immunotherapy or switched to immunotherapy combined with chemotherapy (95).
During the 4 weeks preceding surgery, changes in the density of the TILs were continuously monitored. Patients with increases in the number of CD8+ TILs of ≥30% were scheduled for surgery as planned, whereas those with less marked increases received either adjuvant RT or targeted therapy. Due to the spatial heterogeneity in tumor biomarker expression (Table VI), intraoperative multi-region sampling (core tumor area and peripheral zone) combined with frozen-section pathology and IHC analysis was performed to stratify patients for risk assessment and guide adjuvant therapy decisions. High-risk patients (who were core MRD+ with peripheral zone immunosuppression) were recommended adjuvant immunotherapy combined with chemotherapy to enhance the clearance of residual lesions in the core area. By contrast, low-risk patients (who were core MRD− with peripheral zone immune activation) were advised to receive adjuvant immunotherapy alone in order to reduce chemotherapy toxicity (96).
Table VI.Microenvironmental, biomarker and clinical differences in tumor core zone and tumor edge zone. |
Thoracic drainage fluid ctDNA testing was performed within 24 h postoperatively. If patients were reported to be positive (tumor fraction ≥0.01%), early adjuvant therapy was initiated within 72 h. Additionally, TME spatial mapping was used to evaluate postoperative margins. If the number of CD8+ TILs were <100 cells/high-power field at the invasive margins, high-risk areas were marked to guide postoperative RT target design. At 4 weeks post-surgery, plasma ctDNA levels could be used to assess the MRD status. If two consecutive tests exhibit negative results, the adjuvant immunotherapy cycle may be shortened from the standard 16 cycles to 8 cycles. Lastly, patients should undergo regular soluble PD-L1 (sPD-L1) level testing every 3 months. If the sPD-L1 level should increase by 2 times or more compared with baseline values, the adjuvant therapy should be switched to a combination of immunotherapy and anti-angiogenesis treatment [since the increase in the sPD-L1 level is notably associated with increased levels of VEGF in the TME (97)].
The combined strategy of perioperative immunotherapy should balance efficacy and safety, particularly considering the impact of treatment on surgical feasibility, postoperative recovery and long-term survival. The following section combines the latest clinical studies to explore the potential and challenges of different combined modes in perioperative therapy. Immunotherapy with dual checkpoint inhibitors, via simultaneously blocking multiple immune suppression pathways, including the PD-1/PD-L1 and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) pathways, enhances the antitumor immune response, thereby helping to overcome drug resistance to a certain extent. This strategy has been validated in each of the Checkmate 227, Checkmate 568 and Checkmate 817 trials for its efficacy and safety in advanced NSCLC (10,98–100) and its application in perioperative care is currently being actively explored.
In the NEOpredict-Lung study (101), patients who received nivolumab plus relatlimab as neoadjuvant therapy had an MPR rate of 30% and a pCR rate of 17%. Patients in the PD-L1 ≥50% subgroup demonstrated both a further pathological remission and a lower proportion of residual tumor cells. By contrast, in the CheckMate 816 study, the MPR rate for patients receiving neoadjuvant nivolumab plus chemotherapy was only 35.4% and the pCR rate was 25.3% (13), demonstrating that the dual immunotherapy regimen was able to achieve similar pathological remission levels without chemotherapy. In the EAST ENERGY trial (89), the dual immunotherapy regimen demonstrated even more notable advantages, with an MPR rate of 50% and a pCR rate of 25% for patients receiving neoadjuvant pembrolizumab plus ramucirumab. Additionally, the dual immunotherapy regimen also demonstrated promising results in terms of surgical feasibility. In the NEOpredict-Lung study, patients who received nivolumab plus relatlimab as neoadjuvant therapy achieved a 100% surgical completion rate and a 95% R0 resection rate, with no delays due to treatment toxicity. By contrast, in the CheckMate 77T study, the surgical completion rate for patients receiving neoadjuvant nivolumab combined with chemotherapy was only 77.7% and certain patients required adjustments to their surgical plans due to bone marrow suppression.
Regarding the safety of the dual immunotherapy regimen, in the NEOpredict-Lung trial, patients receiving nivolumab plus relatlimab neoadjuvant therapy were reported to have only a 13% incidence of TRAEs of grade 3 or higher and experienced no chemotherapy-associated toxicities (such as neutropenia). Additionally, irAEs were mainly associated with thyroid dysfunction, with a 40% incidence rate in the dual immunotherapy group and rare serious events, such as pneumonia, which occurred in <2% of cases. By contrast, in the CheckMate 77T study, the incidence of TRAEs of grade 3 or higher was as high as 32.5%, accompanied by notable chemotherapy-associated toxicities, including a 10.1% incidence of neutropenia. Although the dual immunotherapy regimen was associated with higher levels of pathological remission in the PD-L1 level ≥50% group, it still provided notable benefits to patients with PD-L1 levels <1%; for example, in the NEOpredict-Lung study, the MPR rate for patients with PD-L1 levels <1% was 16.7%. Overall, the dual immunotherapy regimen avoids the risks of bone marrow suppression and gastrointestinal toxicity associated with chemotherapy, reduces the risk of postoperative infections and delayed recovery and is particularly suitable for elderly or poorly tolerated patients.
Furthermore, the EAST ENERGY study demonstrated that blocking ramucirumab with VEGFR-2 antagonists led to a reshaping of the TME, an enhancement of CD8+ T-cell infiltration and a prolongation of the duration of the immune response. However, the double immunization regimen still needs to be investigated in phase III trials for the purpose of directly comparing the double immunization regimen with the combination of chemotherapy and immunotherapy to further clarify its advantages (10,102).
At present, although no separate studies have been published on perioperative immunotherapy combined with targeted therapy for lung cancer, a potential synergistic antitumor mechanism exists between immunotherapy and targeted therapy. The MARIPOSA-2 study (103) evaluated the efficacy of the EGFR-mesenchymal/epithelial transition factor (MET) bispecific antibody amivantamab in combination with chemotherapy and lazertinib in patients with advanced EGFR-mutated NSCLC. The results demonstrated that the median PFS in the combined treatment group reached 8.3 months, the median intracranial PFS was 12.8 months and the ORR was 63%. This data demonstrates that amivantamab combined with targeted therapy and chemotherapy has notable efficacy in treating advanced EGFR-mutated NSCLC, suggesting the translational potential of immunotherapy combined with targeted therapy in perioperative lung cancer treatment. In particular, the enhanced intracranial activity of lazertinib may prevent occult brain metastasis progression during surgery; in addition, lazertinib inhibits the EGFR signaling pathway, thereby reducing the levels of immune-suppressive cytokines such as IL-6 and VEGF, which has the effects of improving the TME and enhancing the synergistic effect with immunotherapy. Furthermore, the CheckMate 370 (104) and TATTON (105) studies also explored the potential of immunotherapy in combination with targeted therapy in advanced NSCLC and the results obtained from these studies also have the potential to be applied to perioperative immunotherapy in lung cancer in the future.
Furthermore, novel immunomodulators target non-PD-1/PD-L1 immune checkpoints to modulate key signaling pathways in the TME, thereby offering a novel direction for perioperative immunotherapy. The NeoTRACK study (106) explored the use of atezolizumab in combination with the novel T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, tiragolimab, during perioperative care. Tiragolimab is a novel monoclonal antibody that targets the TIGIT receptor, which functions as an inhibitory immune checkpoint receptor primarily expressed on the surface of activated T and NK cells. By binding to the ligand CD155, which is highly expressed on the surface of tumor and antigen-presenting cells, TIGIT transmits inhibitory signals, leading to T-cell exhaustion and NK cell dysfunction. However, a synergistic inhibitory effect also exists between TIGIT and the PD-1/PD-L1 pathway. Blocking TIGIT helps relieve the dual inhibition of T cells and NK cells, thereby enhancing the antitumor immune response. In models of PD-1/PD-L1 inhibitor resistance, TIGIT inhibition has been reported to restore the function of exhausted T cells, thereby overcoming immune escape. This research is currently ongoing (106) and may potentially lead to applications in perioperative immunotherapy for lung cancer in the future.
Although perioperative immunotherapy for lung cancer has been reported to be effective and safe in the aforementioned clinical trials, long-term resistance to immunotherapy remains a notable concern, particularly for patients requiring postoperative adjuvant immunotherapy. Generally, the mechanisms of immunotherapy resistance can be categorized into primary and secondary modes of resistance. The core of primary drug resistance lies in the ‘cold’ nature of the TME, characterized by insufficient immune cell infiltration. This is primarily due to disordered tumor vascular structures and defects in the secretion of chemokines such as CXCL9/10, which prevent CD8+ T cells from effectively infiltrating the tumor. Additionally, mutations or epigenetic silencing in genes associated with antigen presentation, such as β-2 microglobulin and transporter associated with antigen processing 1/2, result in the absence of MHC class I molecule expression, thereby creating a ‘blind spot’ for immune recognition and leading to an immunosuppressive state against the tumor cells. Furthermore, this immunosuppressive state is exacerbated by the infiltration of immune-suppressing cells, such as Tregs and MDSCs. These cells create an inhibitory TME by secreting cytokines such as IL-10 and TGF-β and they continue to support tumor cell survival through abnormal activation of the Wnt/β-catenin or PI3K/AKT signaling pathways (107–109). If the tumor evades the initial immune attack, secondary drug resistance will gradually develop. Initially, drug-resistant subclones of the tumor achieve immune escape through mutations in driver genes, such as the EGFR T790M mutation and MET amplification or through epigenetic remodeling that leads to the loss of neoantigens due to DNA hypermethylation. Furthermore, tumor cells enhance glycolysis via the Warburg effect, leading to lactic acid accumulation in the TME, which directly inhibits T-cell function. Additionally, abnormal tryptophan metabolism mediated by IDO1 depletes key amino acids in the local TME, which further disrupts the antitumor immune response. Epigenetic regulation either alters the accessibility of immune-associated genes through chromatin modifications that are mediated via histone deacetylases or dynamically regulates the expression of immune checkpoints such as PD-L1 through non-coding RNA networks, including circular RNA/N6-methyladenosine modifications, thereby forming a multi-dimensional drug resistance barrier. These mechanisms are interlinked, ultimately leading to a decline in the clinical response to immunotherapy (Fig. 3) (110–112).
Offering neoadjuvant dual immunotherapy for lung cancer may be a viable approach to overcome drug resistance in the future. The NEOSTAR study demonstrated that, in patients treated with neoadjuvant nivolumab plus ipilimumab, tumor tissue samples that were analyzed using multi-color immunofluorescence technology exhibited a marked reduction in Treg cell infiltration. By contrast, the density of Tregs in the neoadjuvant nivolumab monotherapy group did not markedly change either prior to or after treatment. These findings suggest that this neoadjuvant dual immunotherapy approach effectively leads to an improvement in the immune rejection state, possibly due to the high expression of CTLA-4 on the surface of Treg cells. A combination of ipilimumab and other CTLA-4 inhibitors may effectively deplete Treg cells, thereby improving drug resistance (113). Furthermore, since the C-C motif chemokine ligand 2-CC chemokine receptor 2 (CCL2-CCR2) signaling axis is key to the functional specialization of MDSCs, clinical studies have reported that CCR2 antagonists are able to reduce infiltration of the MDSCs (114–116). At present, the LCMC3 study is being followed up with novel research aimed at validating the use of CCR2 antagonists in combination with PD-1 inhibitors for the perioperative treatment of lung cancer (114). Additionally, lymphocyte activation gene 3 (LAG-3), which is highly expressed in exhausted T cells, binds to the T cell receptor-CD3 complex, leading to an inhibition of T-cell activation by disrupting the interaction between co-receptors and the Src kinase family member Lck, thereby negatively regulating immune function. Clinical trials have demonstrated that a combination of the LAG-3 inhibitor relatlimab and the checkpoint inhibitor nivolumab is able to reverse T-cell exhaustion (117–119). The team responsible for the NEOSTAR study aims to further explore such combination therapies to improve drug resistance (120).
In conclusion, perioperative immunotherapy has fundamentally reshaped the therapeutic paradigm for resectable NSCLC. The present systematic analysis has revealed that multimodal strategies, particularly the ‘sandwich approach’ (namely, neoadjuvant chemoimmunotherapy to surgery to adjuvant immunotherapy), demonstrate notably increased pathological response parameters (pCR, 24.8–56 vs. ≤24% in the neoadjuvant-only patient cohort) and survival benefits (EFS HR, 0.40–0.59 vs. 0.76–0.79 in the adjuvant-only group). Notably, the present review has established two key advancements: i) By integrating the current traditional biomarkers and emerging biomarkers, a dynamic monitoring model has been proposed to overcome the spatial and temporal heterogeneity of PD-L1 and a biomarker-guided perioperative immunotherapy regimen was also implemented to achieve truly personalized treatment for patients; and ii) a clinical decision pathway for the omission of adjuvant immunotherapy in NSCLC: The sandwich protocol increases irAEs (≥3 irAEs: chemotherapy group, 40 vs. 18%) and we propose a decision process to determine whether adjuvant immunotherapy should be exempted following neoadjuvant immunotherapy combined with chemotherapy.
Three main topics are involved here; first, concerning biomarker optimization, PD-L1 thresholds remain contentious (for example, the NEOTORCH trial demonstrated no EFS association with PD-L1), whereas TMB standardization is impeded by platform variability (for example, by comparing the FoundationOne®CDx tTMB with bTMB thresholds). Second, as far as oncogenic driver limitations are concerned, ICIs exhibit minimal efficacy in EGFR/ALK-altered NSCLC, thereby necessitating the development of novel combination therapies. Third, regarding long-term irAE impacts, immune-associated sequelae (for example, radiation pneumonitis in 10% of the patients in the NICOLAS trial) require extended follow-up to define surgical/quality-of-life trade-offs.
Four key areas require further exploration in terms of the goal of working toward precision perioperative therapy. The first future direction for research is dynamic biomarker integration, whereby multi-omics models (spatial transcriptomics plus ctDNA) will be developed to track real-time immune remodeling. The second area is novel combination strategies; for example, studying target immunosuppressive niches with TGF-β/IDO1 inhibitors and strengthening antigen presentation via the use of HLA-DR agonists (as validated in the LungMate 002 trial responders). Thirdly, future studies should investigate personalized toxicity mitigation, as in the case of target GEO signatures (for example, IFN-γ pathway genes) to predict irAE susceptibility. Lastly, real-world validation studies are necessary in order to establish registries for special populations (for example, elderly/oligometastatic populations) and to refine clinical guidelines (such as the NCCN/ESMO guidelines).
Not applicable.
The present review was supported by the Science and Technology Department of Yunnan Province, China (grant nos. 202401AY070001-147 and 202503AP140033); Kunming Medical University (grant nos. QL-JBGS-14 and 2025S270).
Not applicable.
GL, XM and HL conceived the present review. XM reviewed the literature and drafted the manuscript. HL provided financial support. XM, LL, HZ and ZL reviewed the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Jemal A, Sung H, Kelly K, Soerjomataram I and Bray F: The Cancer Atlas, Fourth Edition. American Cancer Society; Atlanta (GA), USA: pp. 1–5. 2025 | |
|
Oncology Society of Chinese Medical Association, . Chinese Medical Association guideline for clinical diagnosis and treatment of lung cancer (2024 edition). Zhonghua Yi Xue Za Zhi. 104:3175–3213. 2024.(In Chinese). PubMed/NCBI | |
|
Luo G, Zhang Y, Rumgay H, Morgan E, Langselius O, Vignat J, Colombet M and Bray F: Estimated worldwide variation and trends in incidence of lung cancer by histological subtype in 2022 and over time: A population-based study. Lancet Respir Med. 13:348–363. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Detterbeck FC, Boffa DJ, Kim AW and Tanoue LT: The eighth edition lung cancer stage classification. Chest. 151:193–203. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd FA, Stephens RJ, Dunant A, Torri V, Rosell R, Seymour L, et al: Lung adjuvant cisplatin evaluation: A pooled analysis by the LACE Collaborative Group. J Clin Oncol. 26:3552–3559. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Anile M, Diso D, Mantovani S, Patella M, Russo E, Carillo C, Pecoraro Y, Onorati I, De Giacomo T, Rendina EA and Venuta F: Uniportal video assisted thoracoscopic lobectomy: Going directly from open surgery to a single port approach. J Thorac Dis. 6 (Suppl 6):S641–S643. 2014.PubMed/NCBI | |
|
Liu L, Yan Y, Wang Y, Li Z, Yang L, Yu K and Zhao Z: Comparative efficacy and safety of first-line PD-1/PD-L1 inhibitors in immunotherapy for non-small cell lung cancer: A network meta-analysis. Oncol Lett. 29:1572025. View Article : Google Scholar : PubMed/NCBI | |
|
Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al: Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 375:1823–1833. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, Park K, Alexandru A, Lupinacci L, de la Mora Jimenez E, et al: Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 381:2020–2031. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chaft JE, Oezkan F, Kris MG, Bunn PA, Wistuba II, Kwiatkowski DJ, Owen DH, Tang Y, Johnson BE, Lee JM, et al: Neoadjuvant atezolizumab for resectable non-small cell lung cancer: An open-label, single-arm phase II trial. Nat Med. 28:2155–2161. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, Zahurak M, Yang SC, Jones DR, Broderick S, et al: Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 378:1976–1986. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, Felip E, Broderick SR, Brahmer JR, Swanson SJ, et al: Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med. 386:1973–1985. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Spicer JD, Garassino MC, Wakelee H, Liberman M, Kato T, Tsuboi M, Lee SH, Chen KN, Dooms C, Majem M, et al: Neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab compared with neoadjuvant chemotherapy alone in patients with early-stage non-small-cell lung cancer (KEYNOTE-671): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 404:1240–1252. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Heymach JV, Harpole D, Mitsudomi T, Taube JM, Galffy G, Hochmair M, Winder T, Zukov R, Garbaos G, Gao S, et al: Perioperative durvalumab for resectable Non-small-cell lung cancer. N Engl J Med. 389:1672–1684. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Felip E, Altorki N, Zhou C, Csőszi T, Vynnychenko I, Goloborodko O, Luft A, Akopov A, Martinez-Marti A, Kenmotsu H, et al: IMpower010 Investigators. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): A randomised, multicentre, open-label, phase 3 trial. Lancet. 398:1344–1357. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
O'Brien M, Paz-Ares L, Marreaud S, Dafni U, Oselin K, Havel L, Esteban E, Isla D, Martinez-Marti A, Faehling M, et al: Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): An interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol. 23:1274–1286. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Riely GJ, Wood DE, Aisner DL, Loo BW Jr, Axtell AL, Bauman JR, Bharat A, Chang JY, Desai A, Dilling TJ, et al: NCCN Guidelines® insights: Non-small cell lung cancer, version 7.2025. J Natl Compr Canc Netw. 23:354–362. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Spicer JD, Cascone T, Wynes MW, Ahn MJ, Dacic S, Felip E, Forde PM, Higgins KA, Kris MG, Mitsudomi T, et al: Neoadjuvant and adjuvant treatments for early stage resectable NSCLC: Consensus recommendations from the international association for the study of lung cancer. J Thorac Oncol. 19:1373–1414. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sorscher S: Perioperative nivolumab in resectable lung cancer. N Engl J Med. 391:5732024.PubMed/NCBI | |
|
Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z, Huynh TG, Zhao L, Fulton L, Schultz KR, et al: EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in Non-small cell lung cancer: A Retrospective analysis. Clin Cancer Res. 22:4585–4593. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, Zhao S, Das S, Beckermann KE, Ha L, et al: Fatal toxic effects associated with immune checkpoint inhibitors: A systematic review and Meta-analysis. JAMA Oncol. 4:1721–1728. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Sahin TK, Ayasun R, Rizzo A and Guven DC: Prognostic value of Neutrophil-to-Eosinophil ratio (NER) in cancer: A systematic review and Meta-analysis. Cancers (Basel). 16:36892024. View Article : Google Scholar : PubMed/NCBI | |
|
Vitale E, Rizzo A, Santa K and Jirillo E: Associations between ‘Cancer Risk’, ‘Inflammation’ and ‘Metabolic Syndrome’: A scoping review. Biology (Basel). 13:3522024.PubMed/NCBI | |
|
Bas O, Sahin TK, Karahan L, Rizzo A and Guven DC: Prognostic significance of the cachexia index (CXI) in patients with cancer: A systematic review and meta-analysis. Clin Nutr ESPEN. 68:240–247. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Rizzo A, Dall'Olio FG, Altimari A, Giunchi F and Ardizzoni A: Role of PD-L1 assessment in advanced NSCLC: Does it still matter? Anticancer Drugs. 32:1084–1085. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rizzo A, Mollica V, Marchetti A, Nuvola G, Rosellini M, Tassinari E, Molina-Cerrillo J, Myint ZW, Buchler T, Monteiro FSM, et al: Adjuvant PD-1 and PD-L1 inhibitors and Relapse-free survival in cancer patients: The MOUSEION-04 study. Cancers (Basel). 14:41422022. View Article : Google Scholar : PubMed/NCBI | |
|
De Zuani M, Xue H, Park JS, Dentro SC, Seferbekova Z, Tessier J, Curras-Alonso S, Hadjipanayis A, Athanasiadis EI, Gerstung M, et al: Single-cell and spatial transcriptomics analysis of non-small cell lung cancer. Nat Commun. 15:43882024. View Article : Google Scholar : PubMed/NCBI | |
|
Amilo D, Izuchukwu C, Sadri K, Yao HR, Hincal E and Shehu Y: A fractional-order model for optimizing combination therapy in heterogeneous lung cancer: Integrating immunotherapy and targeted therapy to minimize side effects. Sci Rep. 14:184842024. View Article : Google Scholar : PubMed/NCBI | |
|
Munteanu R, Tomuleasa C, Iuga CA, Gulei D and Ciuleanu TE: Exploring therapeutic avenues in lung cancer: The epigenetic perspective. Cancers (Basel). 15:53942023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Jiang T, Fang L, Zhang T, Kang X and Zhou C: A randomized, doubleb linded, multicenter phase 3 study of platinum-based chemotherapy with or without QL1706 as adjuvant therapy in completely resected stage II–IIIb NSCLC. J Clin Oncol. 41:TPS86062023. View Article : Google Scholar | |
|
Calvo V, Domine M, Sullivan I, Gonzalez-Larriba J, Ortega A, Bernabé R, Sala M, Campos B, De Castro J, Martín-Martorell P, et al: A phase III clinical trial of adjuvant chemotherapy versus chemoimmunotherapy for stage IB-IIIA completely resected non-small cell lung cancer (NSCLC) patients Nadim-adjuvant: New adjuvant trial of chemotherapy versus chemoimmunotherapy. J Clin Oncol. 39:TPS85812021. View Article : Google Scholar | |
|
Cascone T, Spira A, Campelo R, Kim D, Hamid O, Soo-Hoo Y, Kumar R, Grenga I and Forde P: Abstract CT124: NeoCOAST-2: A randomized, open-label, phase 2 study of neoadjuvant durvalumab plus novel immunotherapies and chemotherapy (CT) followed by adjuvant durvalumab plus novel agents, in patients with resectable non-small-cell lung cancer (NSCLC). Cancer Res. 82:CT1242022. View Article : Google Scholar | |
|
Besse B, Adam J, Cozic N, Chaput-Gras N, Planchard D, Mezquita L, Masip J, Lavaud P, Naltet C, Gazzah A, et al: Neoadjuvant atezolizumab (A) for resectable non-small cell lung cancer (NSCLC): Results from the phase II PRINCEPS trial. Ann Oncol. 31 (Suppl):S794–S795. 2020. View Article : Google Scholar | |
|
Peters S, Spigel D, Ahn M, Tsuboi M, Chaft J, Harpole D, Goss G, Barlesi F, Abbosh C, Poole L, et al: P03.03 MERMAID-1: A Phase III study of adjuvant durvalumab plus chemotherapy in resected NSCLC patients with MRD Post-surgery. J Thorac Oncol. 16 (Suppl):S258–S259. 2021. View Article : Google Scholar | |
|
Spigel D, Peters S, Ahn M, Tsuboi M, Chaft J, Harpole D, Barlesi F, Abbosh C, Mann H, May R, et al: MERMAID-2: Phase III study of durvalumab in patients with resected, stage II–III NSCLC who become MRD after curative-intent therapy. J Thorac Oncol. 16 (Suppl):S745–S746. 2021. View Article : Google Scholar | |
|
Goss G, Darling GE, Westeel V, Nakagawa K, Massuti Sureda B, Perrone F, McLachlan S-A, Kang JH, Wu Y-L, Dingemans A-M.C..et al: LBA48 CCTG BR.31: A global, double-blind placebo-controlled, randomized phase III study of adjuvant durvalumab in completely resected non-small cell lung cancer (NSCLC). Ann Oncol. 35 (Suppl):S12382024. View Article : Google Scholar | |
|
Chen DS and Mellman I: Oncology meets immunology: The cancer-immunity cycle. Immunity. 39:1–10. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Galluzzi L, Buqué A, Kepp O, Zitvogel L and Kroemer G: Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 17:97–111. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Starzer AM, Preusser M and Berghoff AS: Immune Escape mechanisms and therapeutic approaches in cancer: The cancer-immunity cycle. Ther Adv Med Oncol. 14:175883592210962192022. View Article : Google Scholar : PubMed/NCBI | |
|
Mellman I, Chen DS, Powles T and Turley SJ: The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity. 56:2188–2205. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chu NQ, Tan KS, Dycoco J, Adusumilli PS, Bains MS, Bott MJ, Downey RJ, Gray KD, Huang J, Isbell JM, et al: Determinants of successful minimally invasive surgery for resectable non-small cell lung cancer after neoadjuvant therapy. J Thorac Cardiovasc Surg. 169:753–762.e6. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Miyoshi T and Tsuboi M: Postoperative adjuvant therapy with molecularly targeted agents for non-small cell lung cancer. Int J Clin Oncol. 30:210–214. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Kim HK, Joung JG, Choi YL, Lee SH, Park BJ, Choi YS, Ryu D, Nam JY, Lee MS, Park WY, et al: Earlier-phased cancer immunity cycle strongly influences cancer immunity in operable Never-smoker lung adenocarcinoma. iScience. 23:1013862020. View Article : Google Scholar : PubMed/NCBI | |
|
Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M, Hosoi A, Matsushita H, Morishita Y, Kashiwabara K, et al: An immunogram for the Cancer-immunity cycle: Towards personalized immunotherapy of lung cancer. J Thorac Oncol. 12:791–803. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pio R, Ajona D, Ortiz-Espinosa S, Mantovani A and Lambris JD: Complementing the Cancer-immunity cycle. Front Immunol. 10:7742019. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Pu H, Zhang X, Guo X, Li G and Zhang M: Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol. 209:1046832025. View Article : Google Scholar : PubMed/NCBI | |
|
Huang MY, Jiang XM, Wang BL, Sun Y and Lu JJ: Combination therapy with PD-1/PD-L1 blockade in non-small cell lung cancer: Strategies and mechanisms. Pharmacol Ther. 219:1076942021. View Article : Google Scholar : PubMed/NCBI | |
|
Vansteenkiste J, Wauters E, Park K, Rittmeyer A, Sandler A and Spira A: Prospects and progress of atezolizumab in non-small cell lung cancer. Expert Opin Biol Ther. 17:781–789. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang B, Xiao H, Pu X, Zhou C, Yang D, Li X, Wang W and Xiao Q: A real-world comparison between neoadjuvant chemoimmunotherapy and chemotherapy alone for resectable non-small cell lung cancer. Cancer Med. 12:274–286. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cascone T, William WN Jr, Weissferdt A, Leung CH, Lin HY, Pataer A, Godoy MCB, Carter BW, Federico L, Reuben A, et al: Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: The phase 2 randomized NEOSTAR trial. Nat Med. 27:504–514. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rosner S, Reuss JE, Zahurak M, Zhang J, Zeng Z, Taube J, Anagnostou V, Smith KN, Riemer J, Illei PB, et al: Five-year clinical outcomes after neoadjuvant nivolumab in resectable Non-small cell lung cancer. Clin Cancer Res. 29:705–710. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Yin R and Xu L: Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 379:e142018.PubMed/NCBI | |
|
Kim SS, Cooke DT, Kidane B, Tapias LF, Lazar JF, Awori Hayanga JW, Patel JD, Neal JW, Abazeed ME, Willers H, et al: The society of thoracic surgeons expert consensus on the multidisciplinary management and resectability of locally advanced Non-small cell lung cancer. Ann Thorac Surg. 119:16–33. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Chen KN, Chen Q, Wu L, Wang Q, Li X, Ying K, Wang W, Zhao J, Liu L, et al: Neoadjuvant nivolumab plus chemotherapy versus chemotherapy for resectable NSCLC: Subpopulation analysis of Chinese patients in CheckMate 816. ESMO Open. 8:1020402023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu X, Sun L, Song N, He W, Xie B, Hu J, Zhang J, Yang J, Dai J, Bian D, et al: Safety and effectiveness of neoadjuvant PD-1 inhibitor (toripalimab) plus chemotherapy in stage II–III NSCLC (LungMate 002): An open-label, single-arm, phase 2 trial. BMC Med. 20:4932022. View Article : Google Scholar : PubMed/NCBI | |
|
Wislez M, Mazieres J, Lavole A, Zalcman G, Carre O, Egenod T, Caliandro R, Dubos-Arvis C, Jeannin G, Molinier O, et al: Neoadjuvant durvalumab for resectable non-small-cell lung cancer (NSCLC): Results from a multicenter study (IFCT-1601 IONESCO). J Immunother Cancer. 10:e0056362022. View Article : Google Scholar : PubMed/NCBI | |
|
Lei J, Zhao J, Gong L, Ni Y, Zhou Y, Tian F, Liu H, Gu Z, Huang L, Lu Q, et al: Neoadjuvant camrelizumab plus Platinum-based chemotherapy vs Chemotherapy alone for Chinese patients with resectable stage IIIA or IIIB (T3N2) Non-small cell lung cancer: The TD-FOREKNOW randomized clinical trial. JAMA Oncol. 9:1348–1355. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu SY, Dong S, Yang XN, Liao RQ, Jiang BY, Wang Q, Ben XS, Qiao GB, Lin JT, Yan HH, et al: Neoadjuvant nivolumab with or without platinum-doublet chemotherapy based on PD-L1 expression in resectable NSCLC (CTONG1804): A multicenter open-label phase II study. Signal Transduct Target Ther. 8:4422023. View Article : Google Scholar : PubMed/NCBI | |
|
Shao M, Yao J, Wang Y, Zhao L, Li B, Li L, Wu Z, Chen Z, Fan J and Qiu F: Two vs three cycles of neoadjuvant sintilimab plus chemotherapy for resectable non-small-cell lung cancer: NeoSCORE trial. Signal Transduct Target Ther. 8:1462023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Shao M, Yao J, Zhao L, Li L, Chen M, Zhang Y, Liu H, Chen Z, Li B, et al: NeoSCORE II: Three vs four cycles of neoadjuvant sintilimab + chemotherapy for squamous non-small-cell lung cancer. Future Oncol. 20:121–129. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
National Library of Medicine, . Neoadjuvant Sintilimab or Combined With Chemotherapy for Stage III Driven Gene Mutation Negative Non-small Cell Lung Cancer. https://clinicaltrials.gov/study/NCT04728724August 8–2025 | |
|
Kroemer G, Galluzzi L, Kepp O and Zitvogel L: Immunogenic cell death in cancer therapy. Annu Rev Immunol. 31:51–72. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hamada A, Soh J, Hata A, Nakamatsu K, Shimokawa M, Yatabe Y, Suzuki J, Tsuboi M, Horinouchi H, Sakairi Y, et al: Neoadjuvant concurrent Chemo-Immuno-radiation therapy followed by surgery and adjuvant immunotherapy for resectable stage III N2 NSCLC: Primary results from the SQUAT trial (WJOG 12119L). J Thorac Oncol. 20:1098–1107. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Jabbour SK, Lee KH, Frost N, Breder V, Kowalski DM, Pollock T, Levchenko E, Reguart N, Martinez-Marti A, Houghton B, et al: Pembrolizumab plus concurrent chemoradiation therapy in patients with unresectable, locally advanced, stage III Non-small cell lung cancer: The phase 2 KEYNOTE-799 Nonrandomized Trial. JAMA Oncol. 7:1–9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Peters S, Felip E, Dafni U, Belka C, Guckenberger M, Irigoyen A, Nadal E, Becker A, Vees H, Pless M, et al: Safety evaluation of nivolumab added concurrently to radiotherapy in a standard first line chemo-radiotherapy regimen in stage III non-small cell lung cancer-The ETOP NICOLAS trial. Lung Cancer. 133:83–87. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu YP, Li B, Xu XL and Mao WM: Is there a survival benefit in patients with stage IIIA (N2) Non-small cell lung cancer receiving neoadjuvant chemotherapy and/or radiotherapy prior to surgical resection: A systematic review and Meta-analysis. Medicine (Baltimore). 94:e8792015. View Article : Google Scholar : PubMed/NCBI | |
|
Vokes EE, Herndon JE II, Kelley MJ, Cicchetti MG, Ramnath N, Neill H, Atkins JN, Watson DM, Akerley W, Green MR, et al: Induction chemotherapy followed by chemoradiotherapy compared with chemoradiotherapy alone for regionally advanced unresectable stage III Non-small-cell lung cancer: Cancer and Leukemia Group B. J Clin Oncol. 25:1698–704. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Hino A, Muto S, Shimada Y, Hori S, Isotani S, Nagata M and Horie S: Impact of Cisplatin-induced acute kidney injury on Long-term renal function in patients with solid tumors. Clin Exp Nephrol. 27:506–518. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kessler L, Koo C, Richter CP and Tan X: Hearing loss during chemotherapy: Prevalence, mechanisms, and protection. Am J Cancer Res. 14:4597–4632. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Spicer J, Nair S, Khattak A, Lee J, Brown M, Meehan R, Shariati N, Deng X, Samkari A and Chaft J: The phase 3 INTerpath-002 study design: Individualized neoantigen therapy (INT) V940 (mRNA-4157) plus pembrolizumab vs placebo plus pembrolizumab for resected early-stage non-small-cell lung cancer (NSCLC). Cancer Res. 84:CT281. 2024. View Article : Google Scholar | |
|
Garon EB, Lu S, Goto Y, De Marchi P, Paz-Ares L, Spigel DR, Thomas M, Yang JC, Ardizzoni A, Barlesi F, et al: Canakinumab as adjuvant therapy in patients with completely resected Non-Small-cell lung cancer: Results from the CANOPY-A Double-blind, randomized clinical trial. J Clin Oncol. 42:180–191. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chaft J, Dahlberg S, Gerber D, Oxnard G, Malik S, Simone C, Edelman M, Heymach J, Rudin C and Ramalingam S: Adjuvant nivolumab in resected lung cancers (ANVIL): The newest study in the ALCHEMIST platform. J Clin Oncol. 4:352017. | |
|
National Library of Medicine, . A Study of Tiragolumab Plus Atezolizumab Compared With Placebo Plus Atezolizumab in Participants With Completely Resected Non-small Cell Lung Cancer Who Have Received Adjuvant Platinum-based Chemotherapy (SKYSCRAPER-15). https://clinicaltrials.gov/study/NCT06267001August 8–2025 | |
|
Yue D, Wang W, Liu H, Chen Q, Chen C, Liu L, Zhang P, Zhao G, Yang F, Han G, et al: RATIONALE-315 investigators. Perioperative tislelizumab plus neoadjuvant chemotherapy for patients with resectable non-small-cell lung cancer (RATIONALE-315): An interim analysis of a randomised clinical trial. Lancet Respir Med. 13:119–129. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Lu S, Zhang W, Wu L, Wang W, Zhang P, Fang W, Xing W, Chen Q, Yang L, Mei J, et al: Perioperative toripalimab plus chemotherapy for patients with resectable Non-small cell lung cancer: The neotorch randomized clinical trial. JAMA. 331:201–211. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Provencio M, Serna-Blasco R, Nadal E, Insa A, García-Campelo MR, Casal Rubio J, Dómine M, Majem M, Rodríguez-Abreu D, Martínez-Martí A, et al: Overall survival and biomarker analysis of neoadjuvant nivolumab plus chemotherapy in operable stage IIIA Non-Small-Cell lung cancer (NADIM phase II trial). J Clin Oncol. 40:2924–2933. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Provencio M, Nadal E, González-Larriba JL, Martínez-Martí A, Bernabé R, Bosch-Barrera J, Casal-Rubio J, Calvo V, Insa A, Ponce S, et al: Perioperative nivolumab and chemotherapy in Stage III Non-Small-Cell lung cancer. N Engl J Med. 389:504–513. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
National Library of Medicine, . Neoadjuvant and Adjuvant Therapy Studies of Sindilizumab in Resectable Lung Cancer. https://clinicaltrials.gov/study/NCT05116462August 8–2025 | |
|
National Library of Medicine, . Phase III Study of Envafolimab Versus Placebo Plus Chemotherapy in Resectable Stage III NSCLC. https://clinicaltrials.gov/study/NCT06123754August 8–2025 | |
|
Romero Román A, Campo-Cañaveral de la Cruz JL, Macía I, Escobar Campuzano I, Figueroa Almánzar S, Delgado Roel M, Gálvez Muñoz C, García Fontán EM, Muguruza Trueba I, Romero Vielva L, et al: Outcomes of surgical resection after neoadjuvant chemoimmunotherapy in locally advanced stage IIIA non-small-cell lung cancer. Eur J Cardiothorac Surg. 60:81–88. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yan W, Zhong WZ, Liu YH, Chen Q, Xing W, Zhang Q, Liu L, Ge D, Chen K, Yang F, et al: Adebrelimab (SHR-1316) in combination with chemotherapy as perioperative treatment in patients with resectable Stage II to III NSCLCs: An Open-label, multicenter, phase 1b trial. J Thorac Oncol. 18:194–203. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Expert group consensus on standardized perioperative immunotherapy for non-small cell lung cancer, . Consensus expert group on standardized perioperative immunotherapy for Non-small cell lung cancer: Consensus expert opinion on standardized perioperative immunotherapy for Non-small cell lung cancer. China J Clin Oncol. 51:1–14. 2024. | |
|
Provencio M, Nadal E, Insa A, García Campelo R, Casal J, Dómine M, Massuti B, Majem M, Rodríguez-Abreu D, Martínez-Martí A, et al: Perioperative chemotherapy and nivolumab in non-small-cell lung cancer (NADIM): 5-year clinical outcomes from a multicentre, single-arm, phase 2 trial. Lancet Oncol. 25:1453–1464. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
The Multidisciplinary Diagnosis and Treatment (MDT) Committee of the Chinese Anti-Cancer Association, the Small Cell Lung Cancer Expert Committee of the China Society of Clinical Oncology (CSCO), the Tumor Radiotherapy Committee of the Chinese Anti-Cancer Association, . Expert consensus on radiotherapy combined with immunotherapy for inoperable lung cancer (2024 Edition). J Cancer Prevention Control. 31:1223–1239. 2024. | |
|
Uprety D, Roden AC and Peters S: Adjuvant Immunotherapy should be used in patients with Non-Small cell carcinoma with a pathologic complete response to neoadjuvant immunotherapy. J Thorac Oncol. 20:34–38. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Phillips WJ, Camidge DR and Wheatley-Price P: Adjuvant immunotherapy should not be used in patients with a pathologic complete response to neoadjuvant chemoimmunotherapy. J Thorac Oncol. 20:30–33. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Sun T, Chen Z, Wei K and Tang H: Research progress on predictive biomarkers of immunotherapy efficacy in Non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 27:459–465. 2024.(In Chinese). PubMed/NCBI | |
|
Chen K, Yang F, Shen H, Wang C, Li X, Chervova O, Wu S, Qiu F, Peng D, Zhu X, et al: Individualized tumor-informed circulating tumor DNA analysis for postoperative monitoring of non-small cell lung cancer. Cancer Cell. 41:1749–1762.e6. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Peters S, Oliner KS, L'Hernault A, Ratcliffe M, Madison H, Lai Z, Stewart R, Mann H, Lowery C, Garon EB, et al: Durvalumab with or without tremelimumab in combination with chemotherapy in first-line metastatic non-small-cell lung cancer: Outcomes by tumor mutational burden in POSEIDON. ESMO Open. 10:1050582025. View Article : Google Scholar : PubMed/NCBI | |
|
Budczies J, Kazdal D, Menzel M, Beck S, Kluck K, Altbürger C, Schwab C, Allgäuer M, Ahadova A, Kloor M, et al: Tumour mutational burden: Clinical utility, challenges and emerging improvements. Nat Rev Clin Oncol. 21:725–742. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu F, Fan J, He Y, Xiong A, Yu J, Li Y, Zhang Y, Zhao W, Zhou F, Li W, et al: Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer. Nat Commun. 12:25402021. View Article : Google Scholar : PubMed/NCBI | |
|
Long Y, Jin R and Li H: Mechanistic insights into the immune biomarker of perioperative immune checkpoint inhibitors for non-small cell lung cancer. Transl Lung Cancer Res. 14:2821–2841. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ohara S, Suda K and Tsutani Y: Utility and future perspectives of circulating tumor DNA analysis in Non-Small cell lung cancer patients in the era of perioperative Chemo-immunotherapy. Cells. 14:13122025. View Article : Google Scholar : PubMed/NCBI | |
|
Derraze Y, Hashemi SMS, Ulas EB, Ziesemer KA, Lissenberg-Witte B, Radonic T and Bahce I: Evaluating the significance of combining PD-L1 and TILs as biomarkers in non-small cell lung cancer patients treated with immunotherapy: A systematic review. BJC Rep. 3:652025. View Article : Google Scholar : PubMed/NCBI | |
|
Costantini A, Takam Kamga P, Pons-Tostivint E, Fradin D, Emile JF and Giroux-Leprieur E: Soluble PD-L1 (sPD-L1) as a biomarker of durable response and survival in patients with advanced non-small cell lung cancer (NSCLC) treated with first-line immune checkpoint inhibitors (ICIs). Cancer Immunol Immunother. 74:2942025. View Article : Google Scholar : PubMed/NCBI | |
|
Ready N, Hellmann MD, Awad MM, Otterson GA, Gutierrez M, Gainor JF, Borghaei H, Jolivet J, Horn L, Mates M, et al: First-line nivolumab plus ipilimumab in advanced Non-Small-Cell lung cancer (CheckMate 568): Outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J Clin Oncol. 37:992–1000. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ready NE, Audigier-Valette C, Goldman JW, Felip E, Ciuleanu TE, Rosario García Campelo M, Jao K, Barlesi F, Bordenave S, Rijavec E, et al: First-line nivolumab plus ipilimumab for metastatic non-small cell lung cancer, including patients with ECOG performance status 2 and other special populations: CheckMate 817. J Immunother Cancer. 11:e0061272023. View Article : Google Scholar : PubMed/NCBI | |
|
Paz-Ares LG, Ciuleanu TE, Pluzanski A, Lee JS, Gainor JF, Otterson GA, Audigier-Valette C, Ready N, Schenker M, Linardou H, et al: Safety of First-Line Nivolumab Plus Ipilimumab in Patients With Metastatic NSCLC: A Pooled Analysis of CheckMate 227, CheckMate 568, and CheckMate 817. J Thorac Oncol. 8:79–92. 2023. View Article : Google Scholar | |
|
Schuler M, Cuppens K, Plönes T, Wiesweg M, Du Pont B, Hegedus B, Köster J, Mairinger F, Darwiche K, Paschen A, et al: Neoadjuvant nivolumab with or without relatlimab in resectable non-small-cell lung cancer: A randomized phase 2 trial. Nat Med. 30:1602–1611. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Aokage K, Koyama S, Kumagai S, Nomura K, Shimada Y, Yoh K, Wakabayashi M, Fukutani M, Furuya H, Miyoshi T, et al: Efficacy, safety, and influence on the tumor microenvironment of neoadjuvant pembrolizumab plus ramucirumab for PD-L1-Positive NSCLC: A Phase II Trial (EAST ENERGY). Clin Cancer Res. 30:5584–5592. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Passaro A, Wang J, Wang Y, Lee SH, Melosky B, Shih JY, Wang J, Azuma K, Juan-Vidal O, Cobo M, et al: MARIPOSA-2 Investigators. Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on osimertinib: Primary results from the phase III MARIPOSA-2 study. Ann Oncol. 35:77–90. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Spigel DR, Reynolds C, Waterhouse D, Garon EB, Chandler J, Babu S, Thurmes P, Spira A, Jotte R, Zhu J, et al: Phase 1/2 Study of the Safety and Tolerability of Nivolumab Plus Crizotinib for the First-line treatment of anaplastic lymphoma kinase translocation-positive advanced Non-small cell lung cancer (CheckMate 370). J Thorac Oncol. 13:682–688. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Hartmaier RJ, Markovets AA, Ahn MJ, Sequist LV, Han JY, Cho BC, Yu HA, Kim SW, Yang JC, Lee JS, et al: Osimertinib+ savolitinib to overcome acquired MET-mediated resistance in epidermal growth factor Receptor-Mutated, MET-Amplified Non-small cell lung cancer: TATTON. Cancer Discov. 13:98–113. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Roesch RM, Schnorbach J, Klotz LV, Griffo R, Thomas M, Stenzinger A, Christopoulos P, Allgaeuer M, Schneider M, Schuler M, et al: NeoTRACK trial: Neo adjuvant T i R agolumab, A tezolizumab and C hemotherapy-dissection of IO-efficacy in NSCLC by longitudinal trac K ing-protocol of a non-randomised, open-label, single-arm, phase II study. BMJ Open. 15:e0966172025. View Article : Google Scholar : PubMed/NCBI | |
|
Alsaafeen BH, Ali BR and Elkord E: Resistance mechanisms to immune checkpoint inhibitors: Updated insights. Mol Cancer. 24:202025. View Article : Google Scholar : PubMed/NCBI | |
|
Kalbasi A and Ribas A: Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol. 20:25–39. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Said SS and Ibrahim WN: Cancer resistance to immunotherapy: Comprehensive insights with future perspectives. Pharmaceutics. 15:11432023. View Article : Google Scholar : PubMed/NCBI | |
|
Kluger H, Barrett JC, Gainor JF, Hamid O, Hurwitz M, LaVallee T, Moss RA, Zappasodi R, Sullivan RJ, Tawbi H, et al: Society for immunotherapy of cancer (SITC) consensus definitions for resistance to combinations of immune checkpoint inhibitors. J Immunother Cancer. 11:e0059212023. View Article : Google Scholar : PubMed/NCBI | |
|
He J, Huang Z, Han L, Gong Y and Xie C: Mechanisms and management of 3rd-generation EGFR-TKI resistance in advanced non-small cell lung cancer (Review). Int J Oncol. 59:902021. View Article : Google Scholar : PubMed/NCBI | |
|
Cabanos HF and Hata AN: Emerging insights into targeted Therapy-tolerant persister cells in cancer. Cancers (Basel). 13:26662021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAS, Andrews MC, Sharma P, Wang J, Wargo JA, Pe'er D and Allison JP: Distinct cellular mechanisms underlie Anti-CTLA-4 and Anti-PD-1 checkpoint blockade. Cell. 170:1120–1133.e17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Xu M, Wang Y, Xia R, Wei Y and Wei X: Role of the CCL2-CCR2 signalling axis in cancer: Mechanisms and therapeutic targeting. Cell Prolif. 54:e131152021. View Article : Google Scholar : PubMed/NCBI | |
|
Pienta KJ, Machiels JP, Schrijvers D, Alekseev B, Shkolnik M, Crabb SJ, Li S, Seetharam S, Puchalski TA, Takimoto C, et al: Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic Castration-resistant prostate cancer. Invest New Drugs. 31:760–708. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Noel M, O'Reilly EM, Wolpin BM, Ryan DP, Bullock AJ, Britten CD, Linehan DC, Belt BA, Gamelin EC, Ganguly B, et al: Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 38:800–811. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Overman MJ, Gelsomino F, Aglietta M, Wong M, Limon Miron ML, Leonard G, García-Alfonso P, Hill AG, Cubillo Gracian A, Van Cutsem E, et al: Nivolumab plus relatlimab in patients with previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: The phase II CheckMate 142 study. J Immunother Cancer. 12:e0086892024. View Article : Google Scholar : PubMed/NCBI | |
|
Kelly RJ, Landon BV, Zaidi AH, Singh D, Canzoniero JV, Balan A, Hales RK, Voong KR, Battafarano RJ, Jobe BA, et al: Neoadjuvant nivolumab or nivolumab plus LAG-3 inhibitor relatlimab in resectable esophageal/gastroesophageal junction cancer: A phase Ib trial and ctDNA analyses. Nat Med. 30:1023–1034. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gopal AK, Armand P, Neelapu SS, Bartlett NL, Spurgeon SE, Kuruvilla J, Savage KJ, Leonard JP, Gelb AB, Ahmed N, et al: Nivolumab plus relatlimab for patients with relapsed or progressed B-cell malignancies in RELATIVITY-022. Blood Adv. 9:3383–3394. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Aggarwal V, Workman CJ and Vignali DAA: LAG-3 as the third checkpoint inhibitor. Nat Immunol. 24:1415–1422. 2023. View Article : Google Scholar : PubMed/NCBI |