Abstract
Surgery followed by adjuvant chemotherapy is the standard of care for selected patients with early‐stage or locally advanced non‐small cell lung cancer (NSCLC). However, many of these patients still experience postoperative recurrence at 5 years. At present, peri‐operative treatment methods are emerging to prevent early relapse, such as targeted therapy and immunotherapy. Investigation on predictive biomarkers of responses to adjuvant and neoadjuvant therapies is also continuously ongoing. Immunotherapy represented by immune checkpoint inhibitors (ICIs), either by monotherapy or in combination with chemotherapy, has shown benefit in promoting pathological responses and prolonging survival for patients with NSCLC without oncogenic mutations. Exploratory studies have also provided evidence regarding the selection of patients who benefit from ICI‐based perioperative treatment. This review focuses on the existing data of current clinical trials of adjuvant and neoadjuvant strategies with ICIs in resectable NSCLC, the exploration of predictive biomarkers, and the perspectives and urgent challenges in the future.
Keywords: biomarker, peri‐operative immunotherapy, resectable NSCLC
At present, immunotherapy represented by immune checkpoint inhibitors (ICIs), either by monotherapy or in combination with chemotherapy, has shown benefit in promoting pathological responses and prolonging survival for patients with non‐small cell lung cancer (NSCLC) without oncogenic mutations. Investigation on predictive biomarkers of responses to adjuvant and neoadjuvant therapies is also continuously ongoing. This review focuses on the existing data of current clinical trials of adjuvant and neoadjuvant strategies with ICIs in resectable NSCLC, the exploration of predictive biomarkers, and the perspectives and urgent challenges in the future.
Abbreviations
- 18F‐FDG PET‐CT
Fluorine‐18‐fluorodeoxyglucose positron emission tomography‐computed tomography
- BSC
best supportive care
- CI
confidence interval
- CR
complete response
- ctDNA
circulating tumor DNA
- DFS
disease‐free survival
- EFS
event‐free survival
- EGFR
epidermal growth factor receptor
- HR
hazard ratio
- ICIs
immune checkpoint inhibitors
- LCMC
Lung Cancer Mutation Consortium
- MPR
major pathological response
- MRD
molecular residual disease
- NSCLC
non‐small cell lung cancer
- ORR
objective response rate
- OS
overall survival
- pCR
complete pathological response
- PD‐1
programmed cell death‐1
- PD‐L1
programmed cell death‐ligand 1
- PFS
progression‐free survival
- PR
partial response
- RFS
recurrence‐free survival
- SAKK
Swiss cooperative group for Cancer Research
- TC
tumor cell
- TCR
T‐cell receptor
- TMB
tumor mutation burden
- TME
tumor microenvironment
- TPS
tumor proportion score
1. BACKGROUND
Lung cancer is the leading cause of cancer‐related death worldwide [1], and non‐small cell lung cancer (NSCLC) is the most common type, which accounts for around 85% of lung cancer diagnosis [2]. A large amount of patients are diagnosed with advanced or metastatic NSCLC at the time of diagnosis, and only approximately 25% of these patients have localized disease [3]. The standard strategy for patients with resectable NSCLC is surgery followed by adjuvant platinum‐based chemotherapy, which yields an overall survival (OS) advantage of 5% at 5 years [4, 5]. Nevertheless, approximately 50% of patients with Stage II and more than 60% of patients with Stage IIIA disease die within 5 years, despite the standard postoperative chemotherapy [6]. Therefore, exploring novel therapies and treatment strategies is urgently required to reduce the risk of recurrence and to improve the survival of resectable NSCLC. An example of required therapy is that, in patients with epidermal growth factor receptor (EGFR)‐mutant NSCLC, adjuvant‐targeted therapy would prolong disease‐free survival (DFS) after complete resection, which could provide a promising strategy for this subset of the population [7]. However, in those without oncogene mutations, feasible perioperative strategies still require further exploration.
Neoadjuvant treatment is also considered as a potential approach to improve survival in patients with resectable NSCLC, with down‐staging of the tumor and early control of micrometastases. However, the benefit of preoperative chemotherapy is limited. A meta‐analysis showed an absolute 5% survival benefit at 5 years in patients with Stage IB–IIIA NSCLC compared with surgery alone [8], with no significant difference in the survival rate between preoperative and postoperative chemotherapy [9]. The application of blocking antibodies against programmed cell death‐1 (PD‐1) and programmed cell death‐ligand 1 (PD‐L1), named immune checkpoint inhibitor (ICI) therapy, has revolutionized the treatment of advanced NSCLC without driver gene mutations. Research has shown that ICI monotherapy or ICI in combination with chemotherapy prolongs survival in a subset of patients with advanced NSCLC [10, 11]. This finding has led to a growing interest in evaluating the efficacy and safety of ICIs as a setting of adjuvant and neoadjuvant therapy for resectable NSCLC without driver gene mutations. Preclinical model analysis has indicated an improved therapeutic efficacy of neoadjuvant compared with adjuvant immunotherapy to eradicate distant metastases, with increased tumor‐specific CD8 + T cells in peripheral blood and organs, predicting long‐term survival following surgery [12]. Despite the potential feasibility of neoadjuvant or adjuvant immunotherapy in resectable NSCLC shown in multiple trials [13, 14], the efficacy varies in subpopulations. This has resulted in a major pathological response (MPR) rate of 33%–85%, and a complete pathological response (pCR) rate of 10%–71%. Defining patients who could benefit from neoadjuvant immunotherapy by exploring the predictive factors of the treatment strategy is urgently required.
Current studies searching for biomarkers to predict the efficacy of neoadjuvant and adjuvant immunotherapy, and long‐term survival are still on‐going. Investigations of changes in biomarkers in tissue or in peripheral blood before and after neoadjuvant treatment help to comprehensively determine the course of disease and the efficacy of agents. Predictive factors include the expression levels of PD‐L1, tumor mutation burden (TMB), circulating tumor DNA (ctDNA), specific mutant gene pathways, and tumor microenvironment (TME)‐related biomarkers. Because liquid biopsy is noninvasive and widely used, the detection of biomarkers in peripheral blood has been the focus of attention. In this study, we reviewed (1) existing clinical data from emerging immunotherapy trials in neoadjuvant and adjuvant settings, and (2) biomarkers that could affect the efficacy and survival in patients with resectable NSCLC who receive neoadjuvant and adjuvant immunotherapy.
2. MATERIALS AND METHODS
Multiple searches were performed in PubMed only in the English language from inception to September 2022 for clinical trials that included neoadjuvant or adjuvant immunotherapy in resectable NSCLC. Search terms included “early‐stage NSCLC”, “neoadjuvant immunotherapy”, “adjuvant immunotherapy”, and “biomarker”. Moreover, ClinicalTrials. gov was searched by inserting the words “neoadjuvant immunotherapy in NSCLC” and “adjuvant immunotherapy in NSCLC”. In this review, we decided to cite the most relevant studies after an accurate screening, including completed trials with available results.
3. CLINICAL TRIALS THAT INCLUDED ADJUVANT AND NEOADJUVANT IMMUNOTHERAPY IN PATIENTS WITH RESECTABLE NSCLC
3.1. Neoadjuvant immunotherapy in resectable NSCLC
The benefit of neoadjuvant therapy has been investigated and reported, including a reduction in tumor size and an increased rate of R0 resection. The MPR and pCR rates were found to be promising surrogate endpoints for long‐term survival in studies of neoadjuvant therapy [15, 16]. An MPR is usually defined by ≥90% tumor necrosis in the surgical specimen, and a pCR is defined as no viable tumor cells found in the resected samples. A randomized Phase 3 study showed that the pCR and MPR rates were associated with improved event‐free survival (EFS) with neoadjuvant ICI‐based therapy, and the depth of pathological regression also appeared to be predictive of improved EFS [17]. Other endpoints include recurrence‐free survival (RFS), EFS, and the objective response rate (ORR). Multiple studies have shown a favorable safety profile and durable antitumor responses of ICI therapy compared with standard platinum‐based chemotherapy [18, 19]. Therefore, ICI monotherapy was then initially designed in a neoadjuvant setting in preliminary Phase I/II studies. We review the most representative data, such as the CheckMate159 trial, Lung Cancer Mutation Consortium (LCMC) 3 study, and NEOSTAR study (Table 1).
Table 1.
Trial name/NCT number | Phase | Stage | Treatment | Sample size | MPR | pCR |
---|---|---|---|---|---|---|
CheckMate159 (NCT02259621) | I | I‐IIIA | Nivolumab | 22 | 45% | 10% |
LCMC3 (NCT02927301) | II | IB‐IIIA | Atezolizumab | 101 | 20.4% | 6.8% |
NEOSTAR (NCT03158129) | II | I‐IIIA | Nivolumab ± ipilimumab | 44 | 24% versus 50% | 10% versus 38% |
NADIM (NCT03081689) | II | IIIA | Nivolumab + carboplatin/paclitaxel | 46 | 83% | 63% |
NADIM II (NCT03838159) | II | IIIA | Nivolumab + carboplatin/paclitaxel | 87 | 52% | 36.2% |
SAKK 16/14 (NCT02572843) | II | IIIA | Durvalumab + cisplatin/docetaxel | 67 | 62% | 18% |
CheckMate816 (NCT02998528) | III | IB‐IIIA | Nivolumab + platinum‐doublet chemotherapy/platinum‐doublet chemotherapy alone | 358 | 36.9% versus 8.9% | 24% versus 2.2% |
HCRN LUN17‐321 (NCT03871153) | II | III | Durvalumab + carboplatin/paclitaxel + radiation | 25 | Not reported | Not reported |
NCT04061590 | II | I‐IIIA | Pembrolizumab + cisplatin/pemetrexed | NA | Not reported | Not reported |
IMpower030 (NCT03456063) | III | II–IIIB | Atezolizumab + platinum‐based chemotherapy | 453 | Not reported | Not reported |
Abbreviation: NSCLC, non‐small cell lung cancer.
CheckMate159 was one of the first pilot studies that investigated the efficacy and safety of neoadjuvant ICI monotherapy in patients with Stage I–IIIA (AJCC version 7) resectable NSCLC [20]. The PD‐1 inhibitor nivolumab was administered at a dose of 3 mg/kg of body weight every 2 weeks for 1–2 cycles, approximately 4 weeks before the planned surgery. This trial showed an MPR of 45% (9/20 resected tumors, 95% confidence interval [CI]: 23%–68%). The radiological response rate was not completely parallel to pathological responses, with an ORR of 10%. Newly released data showed no correlation between the MPR and RFS, although at a median follow‐up of 30 months, RFS was not matured (with only five recurrent events out of 20 patients) and the 24‐month RFS rate was 69% (95% CI: 51%–93%) [21]. No treatment‐related surgical delays or any previously unreported toxic events were reported, indicating a favorable safety profile [20]. Another trial, ChiCTR‐OIC‐17013726, which evaluated the safety of neoadjuvant PD‐1 inhibitors as the primary endpoint, reported that 52.5% (21/40) of enrolled patients had treatment‐related adverse events after two doses of sintilimab [22]. Only 10.0% (4/40) of patients experienced Grade 3 or higher treatment‐related adverse events, and one patient had Grade 5 treatment‐related adverse event, which indicated that neoadjuvant sintilimab was well tolerated. In this study, the MPR rate was 40.5% in 37 patients who underwent R0 resection, which was also promising [22].
LCMC3 was a Phase 2 study of the neoadjuvant PD‐L1 inhibitor atezolizumab in patients with Stage IB–IIIB NSCLC (AJCC version 7) in whom two cycles of atezolizumab 1200 mg every 3 weeks was administered before surgery [23]. In a preliminary study, the MPR rate was 20.4% (30/147 patients without EGFR/ALK mutations; 95% CI: 14%–28%) and the pCR rate was 6.8% (10/147; 95% CI: 3%–12%) [23]. The presurgery ORR (RECISTv1.1) was 7% [24], which did not reflect the MPR rate, similar to the results from the CheckMate159 study. The NEOSTAR study was a Phase 2 trial of neoadjuvant nivolumab or nivolumab plus ipilimumab in patients with Stage IA–IIIA NSCLC (AJCC version 7) [25]. An MPR rate of 22% and a pCR rate of 10% (2/21) was observed in 21 (5/21) patients who received R0 resection in the nivolumab arm. The ORR was 19% with nivolumab (4/21 complete response [CR]/partial response [PR]), and was positively associated with the pathological tumor response. Notably, some patients with radiographical stable disease also achieved marked pathological regression, similar to all patients with a CR/PR, which indicated that radiographical assessment might not comprehensively reflect tumor regression. This study also showed higher MPR and pCR rates, and less viable tumors in resected specimens in the nivolumab + ipilimumab arm than with nivolumab alone. Therefore, PD‐1 inhibitor‐based neoadjuvant therapy is promising and merits further investigation on the combination strategies for patients with operable NSCLC.
3.2. Neoadjuvant chemoimmunotherapy in resectable NSCLC
Preclinical studies have shown that chemotherapy may work synergically with ICIs in promoting antitumor responses [26]. The mechanistic rationale is that chemotherapy inhibits the proliferation of tumor cells and also exerts immunostimulatory effects by increasing T‐cell infiltration or reducing the immunosuppressive tumor microenvironment [27]. The efficacy of combining immunotherapy and standard chemotherapy has been shown by multiple prospective Phase 3 trials in advanced or metastatic NSCLC, with an ideal response rate in patients, regardless of PD‐L1 expression levels [11, 28]. The strategy of chemoimmunotherapy is more promising for a larger population, with a controllable safety profile, than ICI monotherapy which benefits a subset of patients with high PD‐L1 expression [10, 29]. Phase 1–2 studies have shown that monotherapy of neoadjuvant ICI has an encouraging MPR, which varies from 14% to 40.5% [22, 30]. Additionally, the combination of chemotherapy and an ICI in the neoadjuvant setting is emerging as a novel strategy. The efficacy outcomes from the Phase 2 NADIM study, Swiss cooperative group for Cancer Research (SAKK) 16/14 study, and phase 3 CheckMate 816 study are shown in Table 1.
The NADIM trial assessed the safety and feasibility of the neoadjuvant nivolumab plus paclitaxel‐carboplatin regimen for three cycles before resection, followed by adjuvant treatment with nivolumab until month 12, in 46 patients with stage IIIA (AJCC version 7) NSCLC [31]. The ORR was 76% in 46 patients (2 CRs and 33 PRs), and the progression‐free survival (PFS) rate at 24 months was 77.1%. A total of 89% (41/46) of the study population underwent R0 resection, without any treatment interruption or delay in surgery, and an MPR was achieved in 83% (34/41) of patients, and a pCR was achieved in 63% of patients (26/41). Long‐term follow‐up has also shown a consistent benefit in survival [32, 33]. Notably, the 24‐month PFS rate was 88.4% in 34 patients who achieved an MPR/pCR, 96.2% in 26 who achieved a pCR, and 57.1% in patients with an incomplete pathological response. A significant difference was identified in PFS and OS between patients with a pCR and those with an incomplete pathological response, but not between patients with an MPR and those with an incomplete pathological response [32]. This finding indicated the importance of increasing the proportion of patients with resectable NSCLC who achieve a pCR.
The NADIM II study (NCT03838159) showed a significantly higher pCR rate in the neoadjuvant chemoimmunotherapy group than in the chemotherapy group (36.2% vs. 6.8%, p = 0.0071), and also an improved MPR (52% vs.14%) and ORR (74% vs. 48%) [34]. After a median follow‐up of 26.1 months, the PFS rate at 12 and 24 months reached 89.3% and 66.6% in the chemoimmunotherapy group, which was superior to the chemotherapy group (60.7% and 42.3%), respectively [35]. Additionally, the median PFS was not reached and was 18.3 months in each group (hazard ratio [HR] = 0.48, 95% CI: 0.25–0.91, p = 0.025). The OS rate at 12 and 24 months was 98.2% and 84.7% in the chemoimmunotherapy group, and 82.1% and 63.4% in the chemotherapy group, respectively, and the median OS was not reached in either group (HR = 0.40, 95% CI: 0.17–0.93, p = 0.034) [35]. Overall, neoadjuvant chemoimmunotherapy has benefited the response rate and long‐term survival, which provides more evidence for its application in the perioperative setting.
The SAKK 16/14 study included patients with Stage IIIA (AJCC version 7) NSCLC who were treated with neoadjuvant cisplatin and docetaxel every 3 weeks for two cycles, followed by two cycles of the PD‐L1 inhibitor durvalumab every 2 weeks preoperatively, and continued for 1 year post‐peratively [36]. This strategy led to a further increase in the pathological response rate compared with previous studies of neoadjuvant ICI alone. Among 68 patients enrolled, 55 underwent surgical resection after neoadjuvant sequential chemoimmunotherapy, of whom 34 (62%) achieved an MPR of 62% (34/55) and a pCR of 18% (10/55) [37]. With a median follow‐up of 28.6 months, the EFS at 1 year was 73%, while the median EFS and OS were not reached.
The CheckMate 816 study was the first randomized Phase 3 trial to evaluate the clinical outcome of neoadjuvant chemoimmunotherapy versus chemotherapy as neoadjuvant treatment for Stage IB ( ≥ 4 cm)–IIIA (AJCC version 7) NSCLC [38]. Nivolumab (360 mg) plus platinum‐doublet chemotherapy or chemotherapy alone was administered every 3 weeks for three cycles before planned surgery, followed by up to four cycles of adjuvant chemotherapy, radiotherapy, or both. The baseline characteristics were generally similar in both groups (each with 179 patients), with >60% patients diagnosed with stage IIIA disease, and approximately 50% of patients had pretreatment tumor PD‐L1 expression ≥1% in each group. The median EFS was 31.6 months with chemoimmunotherapy and 20.8 months with chemotherapy alone (p = 0.005) [39]. The pCR rate was 24.0% with nivolumab plus chemotherapy in all of the patients in the primary analysis population, and this was significantly higher than that with chemotherapy alone (2.2%). However, this result was not as remarkable as data from previous sing‐arm Phase 2 studies [31, 37]. A growing number of clinical trials intend to further determine the role of single‐agent immunotherapy or combination therapy in the neoadjuvant setting (Table 1). Although there have been no reports comparing neoadjuvant chemoimmunotherapy and immunotherapy alone, the response rates in current studies appeared to favor the combination strategy. Therefore, currently, there is interest in neoadjuvant chemoimmunotherapy regarding the study design and clinical practice.
3.3. Adjuvant immunotherapy in resectable NSCLC
Adjuvant platinum‐based chemotherapy has been the standard of care for resected stage IB–IIIA NSCLC in past years, with an improvement in OS of 4%–5% 5 years compared with observation [5, 40]. The primary goal of adjuvant therapy is to treat micrometastatic disease as early as possible after surgery to prevent recurrence or distant metastasis. In recent years, the investigation of adjuvant targeted therapy in early stage (Stage IB–IIIA) patients harboring EGFR‐activating mutations has shown a promising benefit of DFS compared with placebo [7]. However, a novel adjuvant treatment for resected NSCLC without EGFR/ALK mutations is still urgently required. Some trials are ongoing, such as the ANVIL study, which randomizes patients with Stage IB–IIIA NSCLC without EGFR/ALK mutations to receive 1 year of nivolumab or observation post‐resection (Table 2).
Table 2.
Trial name/NCT number | Phase | Stage | Regimen | Estimated sample Size | Primary endpoints | Median PFS |
---|---|---|---|---|---|---|
IMpower010 (NCT02486718) | III | IB‐IIIA | Atezolizumab | 1280 | DFS | 42.3 months |
ANVIL (NCT02595944) | III | IB‐IIIA | Nivolumab | 903 | DFS and OS | Not reported |
PEARLS (NCT02504372) | II | IB‐IIIA | Pembrolizumab | 1177 | DFS | Not reported |
BTCRC LUN18‐ 153 (NCT04317534) | II | I | Pembrolizumab | NA | DFS | Not reported |
NCT04585477 | II | I‐III | Durvalumab | 80 | Decrease in ctDNA level | Not reported |
BR31 (NCT02273375) | III | IB‐IIIB | Durvalumab | 1415 | DFS | Not reported |
Abbreviation: NSCLC, non‐small cell lung cancer.
IMpower010 was the first Phase 3 randomized study that showed a promising benefit of DFS with atezolizumab versus best supportive care (BSC) after adjuvant chemotherapy in patients with resected Stages IB–IIIA (AJCC version 7) NSCLC [14]. One to four cycles of platinum‐based chemotherapy were administered in 1280 patients, followed by 1200 mg atezolizumab for up to 16 cycles (or 1 year) or BSC, after randomization stratified on the basis of PD‐L1 expression. The primary endpoint DFS was tested hierarchically in three populations. [41] The first population was the PD‐L1 expression tumor cell (TC) ≥ 1% subgroup with Stage II–IIIA disease, the second was randomized patients with stage II‐IIIA disease, and the third was the intention‐to‐treat (ITT) population, defined as all randomized patients with Stage IB–IIIA disease. Initial data at a median follow‐up of 32.2 months met the primary endpoints. The DFS of adjuvant atezolizumab was significantly longer than that of BSC in patients with Stages II–IIIA and PD‐L1 TC ≥ 1% and in those with stage II–IIIA, with a favorable safety profile with no new safety signals [41]. Based on these results, adjuvant atezolizumab was officially recommended for PD‐L1 TC ≥ 1% in NSCLC without EGFR‐sensitizing mutations in guidelines. Further analysis of the benefit of OS and predictive biomarkers is still undergoing, whereas the application of PD‐L1 inhibitors after adjuvant chemotherapy in early‐stage NSCLC is promising. The clinical benefit of 1%–49% and ≥50% in patients with PD‐L1 expression is shown below to review the predictive value of the PD‐L1 status.
Another Phase 3 study called the PERALS/KEYNOTE‐091 trial compared the efficacy and safety of pembrolizumab and placebo as adjuvant therapy after complete resection in Stage IB–IIIA (AJCC version 7) NSCLC [42]. An interim study showed that adjuvant pembrolizumab prolonged DFS compared with placebo (53.6 vs. 42.0 months, HR = 0.76, 95% CI: 0.63–0.91; p = 0.0014) in the overall population [43]. PD‐L1 testing by an immunohistochemistry 22C3 assay was performed with resected tumors. Adjuvant chemotherapy was considered for stage IB patients and recommended for Stage II–IIIA patients. A subgroup analysis showed that pembrolizumab benefitted DFS in patients who received adjuvant chemotherapy (HR = 0.73 in patients who received chemotherapy and HR = 1.25 in those who did not) [44]. This study provided further evidence for the application of PD‐1 inhibitors in the adjuvant setting for patients with Stage IB–IIIA NSCLC after complete resection and adjuvant chemotherapy if indicated by guidelines.
4. PREDICTIVE BIOMARKERS FOR ADJUVANT AND NEOADJUVANT IMMUNOTHERAPY
4.1. PD‐L1/TMB
The ligand of immune checkpoint receptor PD‐1 is expressed in immune cells and tumor cells, and is highly expressed on the surface of activated lymphocytes, thus inducing suppression of effector immune responses [45]. Therefore, ICIs targeting the PD‐1 pathway may enhance antitumor responses by diminishing suppressive activity in T cell activation within tumors. The utility of PD‐L1 as a biomarker of response to first‐line PD‐1/PD‐L1 inhibitors in NSCLC is widely accepted. In patients with metastatic NSCLC, tumor PD‐L1 expression levels predict responses and outcomes to anti‐PD‐L1 treatment [46, 47] The TMB is also an emerging biomarker in immunotherapy in advanced or metastatic NSCLC. The TMB is a measurement of the number of somatic mutations via next‐generation sequencing derived from tissue or peripheral blood. A high TMB status increases the probability of immune recognition and killing tumor cells, and may indicate an ICI response and clinical outcomes [48]. Despite the theoretical benefit, previous studies have shown controversial results regarding the correlation of TMB and outcomes in patients treated with ICIs. A high tissue TMB status was considered to be correlated with favorable tumor response to pembrolizumab monotherapy in the KEYNOTE‐158 study with previously treated solid tumors [49]. Additionally, a benefit of long‐term survival was observed in patients with advanced NSCLC who received nivolumab plus ipilimumab as the first‐line therapy, regardless of the TMB status in the CheckMate227 study [50]. An updated version of the NCCN guidelines has removed TMB as an emerging immune biomarker for patients with metastatic NSCLC because of the lack of supporting data and concerns of challenges in standardizing laboratory measurements. The investigation of TMB in conjunction of PD‐L1 expression is still on‐going in patients with resectable NSCLC who receive adjuvant and neoadjuvant immune‐based therapies.
With neoadjuvant immunotherapy emerging as a novel strategy in recent years, the predictive role of PD‐L1 to responses of neoadjuvant ICI is also under investigation. The MPR has been accepted as a surrogate endpoint for evaluating the feasibility of immunotherapy in the neoadjuvant setting. Therefore, the relationship between pretreatment/posttreatment PD‐L1 expression and the MPR rate has been investigated in multiple studies. The results of the CheckMate159 trial were not different regarding the MPR between PD‐L1‐positive and PD‐L1‐negative tumors [20]. A relatively small sample size of pretreatment biopsy underwent PD‐L1 expression testing by the Dako 28‐8 assay. Therefore, the evidence provided by subgroup analysis did not appear to be strong enough. However, a significant correlation between the MPR and pretreatment TMB in 11 resected specimens using whole‐exome sequencing in this trial was observed, but there was no significant correlation between TMB and PD‐L1 expression. Interim analysis of biomarkers in the LCMC3 study showed no correlation between the MPR and TMB rates or tumor PD‐L1 expression [51]. Nevertheless, newly released data from the LCMC3 study have shown a positive association between the MPR and PD‐L1 status (clone 22C3), and also showed a positive correlation between the TMB and pathological response [52]. A phase 1b trial examined the safety and efficacy of the neoadjuvant PD‐1 inhibitor sintilimab in 40 patients with Stage IA–IIIB NSCLC [22]. This trial showed a positive correlation between baseline PD‐L1 expression in stromal cells and pathological regression (p = 0.0471), which provides a novel aspect of underlying mechanisms.
The NEOSTAR study showed no association between posttherapy tumor PD‐L1 expression by immunohistochemistry and the pathological response, and a radiographic or pathological response was observed in patients with pretreatment tumor specimens lacking PD‐L1 expression [25]. Similarly, no significant association was observed between pretreatment PD‐L1 expression and the MPR or EFS at 1 year in a multicenter, single‐arm, Phase 2 trial (SAKK16/14), despite a higher pCR rate in patients with PD‐L1 expression levels ≥25% [37]. Moreover, there was no association between the TMB and EFS, MPR, or nodal clearance in another study [53]. In a molecular analysis in the NADIM and NADIM II studies, the PD‐L1 tumor proportion score was significantly higher in patients who had a pCR than in those with an incomplete pathological response [31, 34]. Additionally, increasing categories of PD‐L1 tumor proportion score (TPS) showed an increase in the pCR rate (TPS < 1%, pCR: 14.3%; TPS of 1%–49%, pCR: 41.7%; TPS ≥ 50%, pCR: 61.1%; p = 0.008). However, no similar association was observed in those with an MPR and an incomplete pathological response, or between the PD‐L1 tumor proportion score and PFS or OS [31]. A subgroup analysis of the CheckMate 816 study showed a benefit in the pCR with neoadjuvant nivolumab plus chemotherapy, regardless of tumor PD‐L1 expression levels [39]. However, patients with tumor PD‐L1 expression levels of at least 1% appeared to benefit more than those with levels <1%. The predictive value of PD‐L1 expression and TMB is still debatable, as they are in immunotherapy for advanced diseases, and more biomarkers beyond PD‐L1 expression and TMB need to be investigated.
In the adjuvant setting, the correlation between PD‐L1 expression and the efficacy of the PD‐L1 inhibitor atezolizumab was analyzed with the SP263 immunohistochemistry assay in the IMpower010 study [14]. This study met the primary endpoint DFS in patients with Stage II–IIIA and PD‐L1 TC ≥ 1% (HR = 0.66, 95% CI: 0.50–0.88) and the secondary endpoint DFS in patients with Stage II–IIIA and PD‐L1 TC ≥ 50% (HR = 0.43, 95% CI: 0.27–0.68). An exploratory analysis in the Stage II–IIIA population with PD‐L1 TC of 1%–49% also showed a benefit of DFS (HR = 0.87, 95% CI: 0.60–1.26). The safety profile and clinical benefit with atezolizumab were also consistent across most subgroups in an exploratory analysis of the subpopulation of PD‐L1 TC of ≥50% [54]. The KEYNOTE‐091 study showed that the adjuvant PD‐1 inhibitor pembrolizumab benefited patients with Stage IB–IIIA NSCLC regarding DFS, regardless of PD‐L1 expression after stratification by PD‐L1 levels (<1%, 1%–49%, and ≥50%) [43]. Despite these informative findings, further investigation on the prognostic value of PD‐L1 expression on adjuvant immune‐based therapies is required. Moreover, the challenges in defining the threshold of a high TMB and standardizing the sequencing approach in assessing the TMB still need to be taken into consideration.
4.2. ctDNA/MRD
ctDNA shows a genetic profile in tumors and has emerged as a noninvasive liquid biopsy tool. The genomic changes shown by monitoring ctDNA has provided evidence for assessing tumor responses to antitumor therapies and thus helped guide clinical treatments [55, 56]. In patients with early‐stage NSCLC who undergo complete resection, the detection of ctDNA in peripheral blood helps to identify molecular residual disease (MRD), which might develop in distant metastatic sites. The prospective study LUNGCA‐1 included patients with pathological Stage I–III (AJCC version 8) NSCLC and showed that MRD was an independent factor to predict the risk of relapse after resection [57]. Additionally, the presence of postoperative ctDNA‐based MRD was strongly correlated with a higher rate of disease relapse, and patients who were MRD‐positive and received adjuvant therapies had better RFS than their counterparts without adjuvant therapy [7].
In the NADIM study, which showed 69.8% (30/43) of ctDNA in pretreatment plasma samples, showed that a low baseline ctDNA level was an independent predictive factor for PFS and OS after neoadjuvant chemotherapy plus nivolumab (PFS: HR = 0.20, 95% CI: 0.06–0.63, p = 0.006; OS: HR = 0.07, 95% CI: 0.01–0.39, p = 0.002) [58]. In this study, patients also had low ctDNA levels at baseline had a significantly longer PFS and OS after neoadjuvant chemoimmunotherapy than those with high ctDNA levels [59]. These findings suggest the prognostic value of baseline ctDNA, which is consistent with other studies on advanced cancer treated with ICIs in which a correlation between pretreatment ctDNA and long‐term survival was observed [60].
In contrast, the CheckMate 159 trial, which assessed longitudinal molecular data in peripheral blood for ctDNA, showed that the presence of ctDNA at diagnosis was not associated with RFS. Additionally, clearance of detectable ctDNA before surgery was observed in all patients with a reduction ≥30% in the viable tumor in response to nivolumab [21]. This result shows potential of ctDNA monitoring in predicting the efficacy of neoadjuvant immunotherapy. The Phase 3 trial CheckMate816 provided further evidence in the predictive value of ctDNA clearance in neoadjuvant chemotherapy plus nivolumab treatment using whole‐exome sequencing (before and after two cycles of neoadjuvant thearpy [38]. A ctDNA clearance rate of 56% and 35% was observed in the nivolumab plus chemotherapy group (24/43) and the chemotherapy group (15/44), respectively [39]. Additionally, there was a higher pCR rate in patients with ctDNA clearance in both treatment groups than in those in the groups without ctDNA clearance [39]. This informative data suggests that ctDNA clearance reflects the pathological response and EFS in the therapy used. However, because baseline ctDNA can also identify patients at high risk of progression and death, the assessment of baseline ctDNA may be more feasible than longitudinal surveillance of ctDNA during perioperative immunotherapy.
Investigation on the role of ctDNA to identify patients with MRD after resection who might benefit from adjuvant immunotherapy has provided limited evidence. In 600 patients with evaluable ctDNA before adjuvant therapy after surgery, adjuvant atezolizumab resulted in better DFS than BSC (19.1 vs. 7.9 months, HR = 0.61, 95% CI: 0.39–0.94) in the IMpower010 study [61]. After stratification by the ctDNA status, a benefit in DFS was primarily observed in PD‐L1 TC ≥ 1% in the ctDNA‐positive and ctDNA‐negative groups. Despite the absolute increase in DFS benefit shown in the ctDNA‐positive subpopulation, there are insufficient data on whether postoperative ctDNA shows a benefit in DFS. Therefore, additional studies using post‐chemotherapy and longitudinal time points are required. There are ongoing biomarker studies on the relevance of ctDNA clearance at several designed time points and early relapse in patients with stage IB–IIIA NSCLC treated with adjuvant chemotherapy plus atezolizumab followed by atezolizumab [62].
4.3. Specific gene mutations
The discovery of driver genes, such as EGFR, ALK, and KRAS, has stimulated research on targeted therapy to these signal pathways [63]. This has to an improvement in the prognosis of the patient subgroup with advanced NSCLC harboring these mutations [64]. While KRAS and BRAF V600E mutations show a superior clinical benefit from immunotherapy [65], multiple studies have shown no long‐term benefit of ICI therapy in patients with driver genes, such as EGFR and ALK alterations [18, 66]. Therefore, first‐line ICIs are recommended only in the subset of EGFR/ALK wild‐type patients with advanced NSCLC. The LCMC4 study is also ongoing and aims to determine the proportion of patients with Stage IA2–III lung cancer who harbor targetable driver genes, such as EGFR mutations and ALK rearrangements for selection of neoadjuvant strategies [67]. Besides the driver genes that are frequently mutated in NSCLC, inhibitory mutations, such as STK11 and KEAP1, often co‐occur with the KRAS mutation, and help to define a subgroup of patients who have a poor response and survival to chemoimmunotherapy [68]. The role of driver genes and inhibitors genes in adjuvant and neoadjuvant ICI therapy for early‐stage NSCLC is still under investigation, and the results in perioperative immuno‐based strategies are discussed below.
Biomarker analysis in the LCMC3 study suggested that patients with STK11/LKB1 and KEAP1 were more likely to be pathological non‐responders to neoadjuvant ICI [24]. Similarly, the NADIM study showed a shorter PFS after neoadjuvant chemoimmunotherapy in patients with STK11, KEAP1, RB1, and EGFR mutations (n = 9) than in those without these mutations (n = 20) [31]. The absence of these mutations appeared to be prognostic for neoadjuvant immunotherapy rather than predictive of the pathological response. However, a subgroup analysis in the IMpower010 study showed a similar benefit of DFS from adjuvant atezolizumab in patients with Stage II–IIIA and PD‐L1 TC ≥ 1% who were EGFR‐positive, EGFR‐negative, or an unknown status [14]. This finding may have been due to the small number of patients with a positive EGFR status (n = 43). When patients with known EGFR/ALK mutations were excluded, the HRs for DFS improved in all PD‐L1 subgroups, except for TC ≥ 50% [61]. The evidence from the subgroups is limited owing to the relatively small sample size. Therefore, whether molecular biomarkers can be used before perioperative immunotherapy in expert consensus remains controversial [69].
4.4. TME‐related biomarkers
The investigation of TME‐related biomarkers has shown predictive value of PD‐L1 expression, tumor‐infiltrating lymphocytes, and T‐cell receptor (TCR) in response to immunotherapy. In perioperative therapy, the monitoring of TME‐related biomarkers via pretreatment and posttreatment tissues and blood samples has shown the potential to predict a therapeutic response. However, whether the dynamic changes in tumor infiltration of specific T cells and TCR repertoires are correlated with the tumor response to adjuvant and neoadjuvant therapy in early‐stage patients remains controversial. The presentation and recognition of neoantigen peptides by TCR bound to major histocompatibility molecules is essential in the immunogenic process [60]. TCR features of diversity, evenness, and clonality are being explored as markers in ICI therapy. A study showed a correlation between dynamic changes in TCR clonality and acquired resistance after an initial response to PD‐1 inhibitors, which was induced by neoantigen loss, in patients with stage IIB–IV NSCLC [70]. However, there is less evidence regarding the predictive role of intratumoral and peripheral TCR features in neoadjuvant ICI treatments in resected NSCLC.
In the CheckMate159 trial of neoadjuvant nivolumab therapy, TCR sequencing was performed in tumors and peripheral blood at the time of resection in nine patients. A higher TCR clonality was observed in tumors with an MPR than in their counterparts [20]. The surveillance of peripheral T cell expansion to identify responders and non‐responders to PD‐1 blockade has also suggested potential biomarkers for response and survival [21]. Similarly, TCR richness and clonality were greater in resected tumors followed neoadjuvant nivolumab or nivolumab + ipilimumab therapy than in matched, tumor‐adjacent, uninvolved lung samples in the NEOSTAR study [25]. However, the number of tested samples was limited in these studies. TCR repertoire assessment in the SAKK 16/15 study showed its potential in predicting the risk of recurrence after neoadjuvant sequential chemoimmunotherapy with durvalumab in stage IIIA/N2 NSCLC. The TCR evenness in pretreatment peripheral blood samples was associated with the 1‐year EFS rate, and an increased TCR richness in posttreatment tissue was correlated with the 1‐year EFS rate, MPR, and nodal clearance [53]. In the exploration of biomarkers in the NADIM study, TCR repertoires in primary tumors or lymph nodes from pretreatment biopsy tissue and posttreatment resected tissue of 19 patients with NSCLC were analyzed in relation to convergence, diversity, evenness, and clonality [71]. Larger scale studies on TME‐related biomarkers are required to verify the predictive role of these features.
Intratumoral and peripheral immunophenotypes may dynamically change in patients who receive pathological regression after neoadjuvant ICI therapy. In the CheckMate159 study, only a few intratumoral macrophages expressing PD‐L1 were observed in pretreatment tissue, whereas CD8 + and PD‐1+ immune cells were infiltrated in posttreatment samples in a patient who received an MPR from neoadjuvant nivolumab [20]. The LCMC3 study showed that patients who received an MPR from neoadjuvant atezolizumab had a decreased number of late‐activated CD4 + and CD8 + T lymphocytes in peripheral blood [72]. The changes in CD3 + T lymphocytes from pretreatment specimens and posttreatment tumors were significantly greater in patients treated with nivolumab + ipilimumab than in those treated with nivolumab alone in the NEOSTAR study [25]. This result indicated that the dynamic alternation in CD3 + T lymphocytes potentially correlated with a better response to neoadjuvant ICIs. Patients with stage IIIA NSCLC who achieved a pCR after neoadjuvant nivolumab and chemotherapy showed downregulation on the levels of activated peripheral CD4 + T cells and NK cells, and a reduction in PD‐L1 expression on immune cells in the NADIM study [73].
Multiple additional analysis in molecular biomarkers has also indicated the potential of immune cells in predicting the pathological response to neoadjuvant immunotherapy in resected NSCLC. A higher frequency of tumor‐infiltrating lymphocytes and effector memory T cells were found by flow cytometry in resected specimens after nivolumab + ipilimumab than after nivolumab monotherapy in the NEOSTAR study [25], which indicated a better pathological response. However, no further investigation of the value of tumor‐specific T cells was performed in this study. A decrease in T lymphocytes in tissue, except for regulatory and regulatory T cells, was observed after neoadjuvant nivolumab and chemotherapy [31], but no similar trend was found in peripheral mononuclear cells [74]. Immune cell analysis in the NADIM study showed a significantly decreased number of activated CD4 + T cells and NK cells in posttreatment blood samples in patients who achieved a pCR, with no reduction in cytokines (e.g., interleukin [IL]‐2, IL‐15, IL‐6, IL‐13, and IFN‐γ) after neoadjuvant treatment in this subpopulation [74]. Despite the informative findings reviewed above, the determination of cellular and cytokine biomarkers in neoadjuvant immunotherapies still requires further investigation in larger patient cohorts.
4.5. Fluorine‐18‐fluorodeoxyglucose positron emission tomography‐computed tomography parameters
Fluorine‐18‐fluorodeoxyglucose positron emission tomography‐computed tomography (18F‐FDG PET‐CT) scans show potential predictive value in the evaluation of therapeutic response to immuno‐based strategies for patients with advanced NSCLC [75]. In resected NSCLC, elevated fluorine‐18‐fluorodeoxyglucose maximum standardized uptake (SUVmax) values are correlated with an immunosuppressive and poorly immune infiltrated tumor microenvironment, and also indicate poor clinical outcomes following immunotherapy [76]. The predictive role of SUVmax changes in the pathological response to neoadjuvant ICI treatment has not been fully investigated. Gao et al. reported that a reduction in SUVmax of primary lesions after anti‐PD‐1 (sintilimab) treatment was significantly correlated with the MPR rate [22]. However, in the NEOSTAR study, the phenomenon of nodal immune flare [77] has been observed in a few cases. An invasive pathological examination of the flared nodes with an increased SUVmax showed changes in immune cell infiltration instead of malignancy [25]. The evidence for the predictive value of changes in SUVmax by positron emission tomography‐computed tomography remains controversial and requires further confirmation in larger studies.
5. CURRENT CHALLENGES AND FUTURE PERSPECTIVES
The application of immunotherapy has revolutionized the treatment of patients with advanced NSCLC and those with resected NSCLC who have a high risk of recurrence. Perioperative therapies eliminate micrometastases, prevent postoperative recurrence, and lead to long‐term benefits. An increasing number of studies have shown a clinical benefit from neoadjuvant immunotherapy by promoting pathological regression and prolonging postoperative survival. Further investigation on the combination of ICI and chemotherapy or ICI dual therapy in the neoadjuvant setting has shown a high rate of pathological responses. Our article is the most up‐to‐date review of the primary results of represented trials and of the potential markers for adjuvant and neoadjuvant ICI‐based therapies.
Despite the encouraging results of multiple studies, there are still current challenges for clinical feasibility. Unsolved controversies in the study design of peri‐operative immunotherapy remain. These challenges include the timing for resection after neoadjuvant therapy, the optimal therapeutic mode including the choice of drugs and the application cycle, and the duration of postoperative therapies. A recent Phase 2 study showed that three cycles of neoadjuvant sintilimab plus chemotherapy treatment achieved a higher MPR rate than two cycles in resectable Stage IB–IIIA NSCLC [78]. Moreover, most of the previous studies were Phase 1–2 exploratory trials, with a single cohort and limited sample size. Therefore, the existing data provide limited evidence for the long‐term benefit of ICI‐based therapy in the neoadjuvant setting. The selection of predictive markers is also an issue for perioperative immunotherapy and chemoimmunotherapy. The role of tissue‐based biomarkers (e.g., PD‐L1/TMB) and blood‐based biomarkers (e.g., ctDNA) has been investigated, and large cohort studies are required to provide sufficient evidence. Other potential biomarkers, such as the microbiome, are also under investigation. Collectively, the existing data are promising for adjuvant and neoadjuvant ICI‐based strategy for patients with resectable NSCLC without driver gene mutations, and also suggest multiple potential molecular biomarkers for efficacy and survival.
AUTHOR CONTRIBUTIONS
Ziyi Xu: Conceptualization (equal); methodology (equal); writing—original draft (lead). Zihua Zou: Project administration (supporting); writing—original draft (supporting); writing—review & editing (supporting). Xuezhi Hao: Conceptualization (supporting); project administration (supporting); writing—review & editing (supporting). Puyuan Xing: Project administration (equal); supervision (equal); writing—review & editing (equal). Junling Li: Project administration (lead); supervision (equal); writing—review & editing (equal).
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
ETHICS STATEMENT
Not applicable.
INFORMED CONSENT
Not applicable.
ACKNOWLEDGMENTS
None.
Xu Z, Zou Z, Hao X, Xing P, Li J. Adjuvant and neo‐adjuvant immunotherapy in resectable non‐small cell lung cancer (NSCLC): current status and perspectives. Cancer Innovation. 2023;2:65–78. 10.1002/cai2.49
DATA AVAILABILITY STATEMENT
Data sharing is not applicable – no new data is generated.
REFERENCES
- 1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33. 10.3322/caac.21654 [DOI] [PubMed] [Google Scholar]
- 3. Howlader N, Noone A, Krapcho M, et al. SEER cancer statistics review, 1975–2018. National Cancer Institute; 2021. [Google Scholar]
- 4. Chemotherapy in non‐small cell lung cancer: a meta‐analysis using updated data on individual patients from 52 randomised clinical trials. non‐small cell lung cancer collaborative group. BMJ (Clinical research ed). 1995;311(7010):899–909. 10.1136/bmj.311.7010.899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Arriagada R, Auperin A, Burdett S, et al. Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non‐small‐cell lung cancer: two meta‐analyses of individual patient data. The Lancet. 2010;375(9722):1267–77. 10.1016/S0140-6736(10)60059-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Goldstraw P, Chansky K, Crowley J, Rami‐Porta R, Asamura H, Eberhardt WEE, et al. The IASLC lung cancer staging project: proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11(1):39–51. 10.1016/j.jtho.2015.09.009 [DOI] [PubMed] [Google Scholar]
- 7. Wu YL, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR‐mutated non‐small‐cell lung cancer. N Engl J Med. 2020;383(18):1711–23. 10.1056/NEJMoa2027071 [DOI] [PubMed] [Google Scholar]
- 8. NSCLC Meta‐analysis Collaborative Group . Preoperative chemotherapy for non‐small‐cell lung cancer: a systematic review and meta‐analysis of individual participant data. The Lancet. 2014;383(9928):1561–71. 10.1016/S0140-6736(13)62159-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lim E, Harris G, Patel A, Adachi I, Edmonds L, Song F. Preoperative versus postoperative chemotherapy in patients with resectable non‐small cell lung cancer: systematic review and indirect comparison meta‐analysis of randomized trials. J Thorac Oncol. 2009;4(11):1380–8. 10.1097/JTO.0b013e3181b9ecca [DOI] [PubMed] [Google Scholar]
- 10. Reck M, Rodríguez‐Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus chemotherapy for PD‐L1‐Positive Non‐Small‐Cell lung cancer. N Engl J Med. 2016;375(19):1823–33. 10.1056/NEJMoa1606774 [DOI] [PubMed] [Google Scholar]
- 11. Gandhi L, Rodríguez‐Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic Non‐Small‐Cell lung cancer. N Engl J Med. 2018;378(22):2078–92. 10.1056/NEJMoa1801005 [DOI] [PubMed] [Google Scholar]
- 12. Liu J, Blake SJ, Yong MCR, Harjunpää H, Ngiow SF, Takeda K, et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discovery. 2016;6(12):1382–99. 10.1158/2159-8290.CD-16-0577 [DOI] [PubMed] [Google Scholar]
- 13. Jia XH, Xu H, Geng LY, Jiao M, Wang WJ, Jiang LL, et al. Efficacy and safety of neoadjuvant immunotherapy in resectable nonsmall cell lung cancer: a meta‐analysis. Lung Cancer. 2020;147:143–53. 10.1016/j.lungcan.2020.07.001 [DOI] [PubMed] [Google Scholar]
- 14. Felip E, Altorki N, Zhou C, Csőszi T, Vynnychenko I, Goloborodko O, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB‐IIIA non‐small‐cell lung cancer (IMpower010): a randomised, multicentre, open‐label, phase 3 trial. The Lancet. 2021;398(10308):1344–57. 10.1016/S0140-6736(21)02098-5 [DOI] [PubMed] [Google Scholar]
- 15. Hellmann MD, Chaft JE, William WN, Jr. , Rusch V, Pisters KMW, Kalhor N, et al. Pathological response after neoadjuvant chemotherapy in resectable non‐small‐cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint. Lancet Oncol. 2014;15(1):e42–50. 10.1016/S1470-2045(13)70334-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Waser NA, Adam A, Schweikert B, Vo L, McKenna M, Breckenridge M, et al. 1243P pathologic response as early endpoint for survival following neoadjuvant therapy (NEO‐AT) in resectable non‐small cell lung cancer (rNSCLC): systematic literature review and meta‐analysis. Ann Oncol. 2020;31:S806. 10.1016/j.annonc.2020.08.116 [DOI] [Google Scholar]
- 17. Provencio‐Pulla M, Spicer J, Taube JM, Martin C, Spigel DR, Wang C, et al. Neoadjuvant nivolumab (NIVO) + platinum‐doublet chemotherapy (chemo) versus chemo for resectable (IB–IIIA) non‐small cell lung cancer (NSCLC): association of pathological regression with event‐free survival (EFS) in CheckMate 816. J Clin Oncol. 2022;40(Suppl 17):LBA8511. 10.1200/JCO.2022.40.17_suppl.LBA8511 [DOI] [Google Scholar]
- 18. Borghaei H, Paz‐Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous Non‐Small‐Cell lung cancer. N Engl J Med. 2015;373(17):1627–39. 10.1056/NEJMoa1507643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous‐cell non‐small‐cell lung cancer. N Engl J Med. 2015;373(2):123–35. 10.1056/NEJMoa1504627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. Neoadjuvant PD‐1 blockade in resectable lung cancer. N Engl J Med. 2018;378(21):1976–86. 10.1056/NEJMoa1716078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Reuss JE, Smith KN, Anagnostou V, et al. Neoadjuvant nivolumab in resectable non‐small cell lung cancer: extended follow‐up and molecular markers of response. J Clin Oncol. 2019;37(suppl):8524. 10.1200/JCO.2019.37.15_suppl.8524 [DOI] [Google Scholar]
- 22. Gao S, Li N, Gao S, Xue Q, Ying J, Wang S, et al. Neoadjuvant PD‐1 inhibitor (Sintilimab) in NSCLC. J Thorac Oncol. 2020;15(5):816–26. 10.1016/j.jtho.2020.01.017 [DOI] [PubMed] [Google Scholar]
- 23. Lee J, Chaft J, Nicholas A, et al. Surgical and clinical outcomes with neoadjuvant atezolizumab in resectable stage IB‐IIIB NSCLC: LCMC3 trial primary analysis. J Thorac Oncol. 2021;16(3):S59–61. 10.1016/j.jtho.2021.01.320 [DOI] [Google Scholar]
- 24. Carbone D, Lee J, Kris M, Wistuba I, Kwiatkowski D, Owen D, et al. OA06.06 Clinical/Biomarker data for neoadjuvant atezolizumab in resectable stage IB‐IIIB NSCLC: primary analysis in the LCMC3 study. J Thorac Oncol. 2021;16(3):S115–6. 10.1016/j.jtho.2021.01.294 [DOI] [Google Scholar]
- 25. Cascone T, William WN, Jr. , Weissferdt A, Leung CH, Lin HY, Pataer A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non‐small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nature Med. 2021;27(3):504–14. 10.1038/s41591-020-01224-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Leonetti A, Wever B, Mazzaschi G, Assaraf YG, Rolfo C, Quaini F, et al. Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non‐small cell lung cancer. Drug Resist Updates. 2019;46:100644. 10.1016/j.drup.2019.100644 [DOI] [PubMed] [Google Scholar]
- 27. Heinhuis KM, Ros W, Kok M, Steeghs N, Beijnen JH, Schellens JHM. Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Ann Oncol. 2019;30(2):219–35. 10.1093/annonc/mdy551 [DOI] [PubMed] [Google Scholar]
- 28. Langer CJ, Gadgeel SM, Borghaei H, Papadimitrakopoulou VA, Patnaik A, Powell SF, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non‐squamous non‐small‐cell lung cancer: a randomised, phase 2 cohort of the open‐label KEYNOTE‐021 study. Lancet Oncol. 2016;17(11):1497–508. 10.1016/S1470-2045(16)30498-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non‐small‐cell lung cancer. N Engl J Med. 2015;372(21):2018–28. 10.1056/NEJMoa1501824 [DOI] [PubMed] [Google Scholar]
- 30. Besse B, Adam J, Cozic N, Chaput N, Mercier O. 1215O ‐ SC neoadjuvant atezolizumab (A) for resectable non‐small cell lung cancer (NSCLC): results from the phase II PRINCEPS trial. ESMO. 2020;2020. 10.1016/j.annonc.2020.08.1417 [DOI] [Google Scholar]
- 31. Provencio M, Nadal E, Insa A, García‐Campelo MR, Casal‐Rubio J, Dómine M, et al. Neoadjuvant chemotherapy and nivolumab in resectable non‐small‐cell lung cancer (NADIM): an open‐label, multicentre, single‐arm, phase 2 trial. Lancet Oncol. 2020;21(11):1413–22. 10.1016/S1470-2045(20)30453-8 [DOI] [PubMed] [Google Scholar]
- 32. Provencio M, Nadal E, Insa A, Campelo MRG, Pereiro D, Domine M, et al. OA20.01 long term survival in operable stage iiia nsclc patients treated with neoadjuvant nivolumab plus chemotherapy ‐ nadim study. J Thorac Oncol. 2021;16(10):S883. 10.1016/j.jtho.2021.08.101 [DOI] [Google Scholar]
- 33. Provencio M, Serna‐Blasco R, Nadal E, Insa A, García‐Campelo MR, Casal Rubio J, 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. 2022;40(25):2924–33. 10.1200/JCO.21.02660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Provencio‐Pulla M, Nadal E, Larriba JLG, Martinez‐Marti A, Bernabé R, Bosch‐Barrera J, et al. Nivolumab + chemotherapy versus chemotherapy as neoadjuvant treatment for resectable stage IIIA NSCLC: primary endpoint results of pathological complete response (pCR) from phase II NADIM II trial. J Clin Oncol. 2022;40:8501. 10.1200/JCO.2022.40.16_suppl.8501 [DOI] [Google Scholar]
- 35. Provencio M, Serna R, Nadal E, Glez Larriba JL, Martínez‐Martí A, Bernabé R, et al. PL03. 12 progression free survival and overall survival in NADIM II study. J Thorac Oncol. 2022;17(9):S2–3. 10.1016/j.jtho.2022.07.014 [DOI] [Google Scholar]
- 36. Rothschild SI, Zippelius A, Eboulet EI, Savic Prince S, Betticher D, Bettini A, et al. 1237MO SAKK 16/14: Anti‐PD‐L1 antibody durvalumab in addition to neoadjuvant chemotherapy in patients with stage IIIA (N2) non‐small cell lung cancer (NSCLC) – A multicenter single‐arm phase II trial. Ann Oncol. 2020;31:S803–4. 10.1016/j.annonc.2020.08.110 [DOI] [PubMed] [Google Scholar]
- 37. Rothschild SI, Zippelius A, Eboulet EI, Savic Prince S, Betticher D, Bettini A, et al. SAKK 16/14: durvalumab in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) Non‐Small‐Cell lung Cancer‐A multicenter Single‐Arm phase II trial. J Clin Oncol. 2021;39(26):2872–80. 10.1200/JCO.21.00276 [DOI] [PubMed] [Google Scholar]
- 38. Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Abstract CT003: nivolumab (NIVO) + platinum‐doublet chemotherapy (chemo) vs chemo as neoadjuvant treatment (tx) for resectable (IB‐IIIA) non‐small cell lung cancer (NSCLC) in the phase 3 CheckMate 816 trial. Cancer Res. 2021;81:CT003. 10.1158/1538-7445.AM2021-CT003 [DOI] [Google Scholar]
- 39. Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med. 2022;386(21):1973–85. 10.1056/NEJMoa2202170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd FA, Stephens RJ, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE collaborative group. J Clin Oncol. 2008;26(21):3552–9. 10.1200/JCO.2007.13.9030 [DOI] [PubMed] [Google Scholar]
- 41. Wakelee HA, Altorki NK, Zhou C, Csőszi T, Vynnychenko IO, Goloborodko O, et al. IMpower010: primary results of a phase III global study of atezolizumab versus best supportive care after adjuvant chemotherapy in resected stage IB‐IIIA non‐small cell lung cancer (NSCLC). J Clin Oncol. 2021;39(Suppl 15):8500. 10.1200/JCO.2021.39.15_suppl.8500 [DOI] [Google Scholar]
- 42. O'Brien MER, Hasan B, Dafni U, Menis J, Peters S, De Waele M, et al. EORTC‐ETOP randomized, phase 3 trial with anti‐PD‐1 monoclonal antibody pembrolizumab versus placebo for patients with early stage non‐small cell lung cancer (NSCLC) after resection and standard adjuvant chemotherapy: PEARLS (NCT02504372). J Clin Oncol. 2016;34(Suppl 15):TPS8571. 10.1200/JCO.2016.34.15_suppl.TPS8571 [DOI] [Google Scholar]
- 43. Paz‐Ares L, O'Brien MER, Mauer M, et al. VP3‐2022: pembrolizumab (pembro) versus placebo for early‐stage non‐small cell lung cancer (NSCLC) following complete resection and adjuvant chemotherapy (chemo) when indicated: randomized, triple‐blind, phase III EORTC‐1416‐LCG/ETOP 8‐15 – PEARLS/KEYNOTE‐091 study. Ann Oncol. 2022;33(4):451–3. 10.1016/j.annonc.2022.02.224 [DOI] [Google Scholar]
- 44. O'Brien MER, Paz‐Ares L, Jha N, Dafni U, Oselin K, Havel L, et al. EORTC‐1416‐LCG/ETOP 8‐15 – PEARLS/KEYNOTE‐091 study of pembrolizumab versus placebo for completely resected early‐stage non‐small cell lung cancer (NSCLC): outcomes in subgroups related to surgery, disease burden, and adjuvant chemotherapy use. J Clin Oncol. 2022;40(Suppl 16):8512. 10.1200/JCO.2022.40.16_suppl.8512 [DOI] [Google Scholar]
- 45. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64. 10.1038/nrc3239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD‐L1‐expressing, locally advanced or metastatic non‐small‐cell lung cancer (KEYNOTE‐042): a randomised, open‐label, controlled, phase 3 trial. The Lancet. 2019;393(10183):1819–30. 10.1016/S0140-6736(18)32409-7 [DOI] [PubMed] [Google Scholar]
- 47. Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated non‐small‐cell lung cancer (POPLAR): a multicentre, open‐label, phase 2 randomised controlled trial. The Lancet. 2016;387(10030):1837–46. 10.1016/S0140-6736(16)00587-0 [DOI] [PubMed] [Google Scholar]
- 48. Sholl LM, Hirsch FR, Hwang D, Botling J, Lopez‐Rios F, Bubendorf L, et al. The promises and challenges of tumor mutation burden as an immunotherapy biomarker: a perspective from the international association for the study of lung cancer pathology committee. J Thorac Oncol. 2020;15(9):1409–24. 10.1016/j.jtho.2020.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Marabelle A, Fakih M, Lopez J, Shah M, Shapira‐Frommer R, Nakagawa K, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open‐label, phase 2 KEYNOTE‐158 study. Lancet Oncol. 2020;21(10):1353–65. 10.1016/S1470-2045(20)30445-9 [DOI] [PubMed] [Google Scholar]
- 50. Hellmann MD, Paz‐Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus ipilimumab in advanced Non‐Small‐Cell lung cancer. N Engl J Med. 2019;381(21):2020–31. 10.1056/NEJMoa1910231 [DOI] [PubMed] [Google Scholar]
- 51. Kwiatkowski DJ, Rusch VW, Chaft JE, et al. Neoadjuvant atezolizumab in resectable non‐small cell lung cancer (NSCLC): interim analysis and biomarker data from a multicenter study (LCMC3). J Clin Oncol. 2019;37(Suppl 15):8503. 10.1200/JCO.2019.37.15_suppl.8503 [DOI] [Google Scholar]
- 52. Chaft JE, Oezkan F, Kris MG, Bunn PA, Wistuba II, Kwiatkowski DJ, et al. Neoadjuvant atezolizumab for resectable non‐small cell lung cancer: an open‐label, single‐arm phase II trial. Nature Med. 2022;28(10):2155–61. 10.1038/s41591-022-01962-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Alborelli I, Leonards K, Manzo M, et al. MA09.02 SAKK 16/14 ‐ T‐cell receptor repertoire metrics predict response to neoadjuvant durvalumab in patients with stage IIIA(N2) NSCLC. J Thorac Oncol. 2021;16(10):S911. 10.1016/j.jtho.2021.08.153 [DOI] [Google Scholar]
- 54. Felip E, Altorki NK, Zhou C, Vallieres E, Vynnychenko IO, Akopov A, et al. 80O Atezolizumab (atezo) vs best supportive care (BSC) in stage II‐IIIA NSCLC with high PD‐L1 expression: sub‐analysis from the pivotal phase III IMpower010 study. Ann Oncol. 2022;33:S71. 10.1016/j.annonc.2022.02.090 [DOI] [Google Scholar]
- 55. Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DWY, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4(136):136ra68. 10.1126/scitranslmed.3003726 [DOI] [PubMed] [Google Scholar]
- 56. Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti‐EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532–6. 10.1038/nature11156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Xia L, Mei J, Kang R, et al. Perioperative ctDNA‐Based molecular residual disease detection for Non‐Small cell lung cancer: a prospective multicenter cohort study (LUNGCA‐1). Clin Cancer Res. 2022;28(15):3308–17. 10.1158/1078-0432.CCR-21-3044 [DOI] [PubMed] [Google Scholar]
- 58. Provencio M, Serna‐Blasco R, Nadal E, Insa A, García‐Campelo MR, Casal Rubio J, 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. 2022;40(25):2924–33. 10.1200/JCO.21.02660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Romero A, Serna R, Nadal E, Larriba JLG, Martínez‐Martí A, Bernabé R, et al. MA06. 03 pre‐treatment ctDNA levels significantly predicts of OS and PFS in NADIM II trial. J Thorac Oncol. 2022;17(9):S63. 10.1016/j.jtho.2022.07.106 [DOI] [Google Scholar]
- 60. Zhang Q, Luo J, Wu S, Si H, Gao C, Xu W, et al. Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discovery. 2020;10(12):1842–53. 10.1158/2159-8290.CD-20-0047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Zhou C, Das Thakur M, Srivastava MK, Zou W, Xu H, Ballinger M, et al. 2O IMpower010: biomarkers of disease‐free survival (DFS) in a phase III study of atezolizumab (atezo) vs best supportive care (BSC) after adjuvant chemotherapy in stage IB‐IIIA NSCLC. Ann Oncol. 2021;32:S1374. 10.1016/j.annonc.2021.10.018 [DOI] [Google Scholar]
- 62. Ardeshir‐Larijani F, Mohindra NA, Traynor AM, Malhotra J, Shum E, Ma PC, et al. Assessing the predictive value of ctDNA on relapse in patients with resected stage IB‐IIIA NSCLC treated with adjuvant chemotherapy plus concomitant atezolizumab followed by atezolizumab: BTCRC LUN 19‐396. J Clin Oncol. 2022;40(Suppl 16):TPS8592. 10.1200/JCO.2022.40.16_suppl.TPS8592 [DOI] [Google Scholar]
- 63. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. 10.1016/j.cell.2011.02.013 [DOI] [PubMed] [Google Scholar]
- 64. Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR‐mutated advanced NSCLC. N Engl J Med. 2020;382(1):41–50. 10.1056/NEJMoa1913662 [DOI] [PubMed] [Google Scholar]
- 65. Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, et al. Oncogene‐specific differences in tumor mutational burden, PD‐L1 expression, and outcomes from immunotherapy in non‐small cell lung cancer. J Immunother Cancer. 2021;9(8):e002891. 10.1136/jitc-2021-002891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Herbst RS, Baas P, Kim DW, Felip E, Pérez‐Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD‐L1‐positive, advanced non‐small‐cell lung cancer (KEYNOTE‐010): a randomised controlled trial. The Lancet. 2016;387 (10027):1540–50. 10.1016/S0140-6736(15)01281-7 [DOI] [PubMed] [Google Scholar]
- 67. Sepesi B, Jones DR, Meyers BF, Chaft JE, Sholl LM, Shyr Y, et al. LCMC LEADER neoadjuvant screening trial: LCMC4 evaluation of actionable drivers in early‐stage lung cancers. J Clin Oncol. 2022;40(Suppl 16):TPS8596. 10.1200/JCO.2022.40.16_suppl.TPS8596 [DOI] [Google Scholar]
- 68. Skoulidis F, Arbour KC, Hellmann MD, Patil PD, Marmarelis ME, Awad MM, et al. Association of STK11/LKB1 genomic alterations with lack of benefit from the addition of pembrolizumab to platinum doublet chemotherapy in non‐squamous non‐small cell lung cancer. J Clin Oncol. 2019;37(Suppl 15):102. 10.1200/JCO.2019.37.15_suppl.102 [DOI] [Google Scholar]
- 69. Liang W, Cai K, Chen C, Chen H, Chen Q, Fu J, et al. Expert consensus on neoadjuvant immunotherapy for non‐small cell lung cancer. Transl Lung Cancer Res. 2020;9(6):2696–715. 10.21037/tlcr-2020-63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, et al. Evolution of neoantigen landscape during immune checkpoint blockade in Non‐Small cell lung cancer. Cancer Discovery. 2017;7(3):264–76. 10.1158/2159-8290.CD-16-0828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Casarrubios M, Nadal E, Cruz‐Bermudez A, Insa A, Briviesca RL, Campelo MRG, et al. P60.11 TCR repertoire predicts pathological response in NSCLC patients receiving neoadjuvant chemoimmunotherapy from NADIM trial. J Thorac Oncol. 2021;16(3):S545–6. 10.1016/j.jtho.2021.01.970 [DOI] [Google Scholar]
- 72. Oezkan F, He K, Owen D, Pietrzak M, Cho JH, Kitzler R, et al. OA13. 07 neoadjuvant atezolizumab in resectable NSCLC patients: immunophenotyping results from the interim analysis of the multicenter trial LCMC3. J Thorac Oncol. 2019;14(10):S242–3. 10.1016/j.jtho.2019.08.482 [DOI] [Google Scholar]
- 73. Laza‐Briviesca R, Cruz‐Bermúdez A, Casarrubios M, Nadal E, Insa A, Garcia Campelo R, et al. P2.04‐10 Biomarkers of pathological response on neo‐adjuvant chemo‐immunotherapy treatment for resectable stage IIIA NSCLC patients. J Thorac Oncol. 2019;14(10):S711. 10.1016/j.jtho.2019.08.1515 [DOI] [Google Scholar]
- 74. Laza Briviesca R, Nadal E, Casarrubios M, Insa A, Sierra‐Rodero B, Garcia Campelo MR, et al. FP12.09 molecular insight into NADIM clinical trial: potential immune biomarkers of pathological response for NSCLC patients. J Thorac Oncol. 2021;16(3):S220–1. 10.1016/j.jtho.2021.01.135 [DOI] [Google Scholar]
- 75. Mitchell KG, Amini B, Wang Y, Carter BW, Godoy MCB, Parra ER, et al. 18F‐fluorodeoxyglucose positron emission tomography correlates with tumor immunometabolic phenotypes in resected lung cancer. Cancer Immunol Immunother. 2020;69(8):1519–34. 10.1007/s00262-020-02560-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Evangelista L, Cuppari L, Menis J, Bonanno L, Reccia P, Frega S, et al. 18F‐FDG PET/CT in non‐small‐cell lung cancer patients: a potential predictive biomarker of response to immunotherapy. Nucl Med Commun. 2019;40(8):802–7. 10.1097/MNM.0000000000001025 [DOI] [PubMed] [Google Scholar]
- 77. Sepesi B, Godoy M, William W, Vaporciyan A, Lin H, Leung C, et al. P2.04‐90 nodal immune flare (NIF) following neoadjuvant Anti‐PD‐1 and Anti‐CTLA‐4 therapy in non‐small cell lung cancer. J Thorac Oncol. 2019;14(10):S745. 10.1016/j.jtho.2019.08.1595 [DOI] [Google Scholar]
- 78. Qiu F, Fan J, Shao M, Yao J, Zhao L, Zhu L, et al. Two cycles versus three cycles of neoadjuvant sintilimab plus platinum‐doublet chemotherapy in patients with resectable non‐small‐cell lung cancer (neoSCORE): a randomized, single center, two‐arm phase II trial. J Clin Oncol. 2022;40(Suppl 16):8500. 10.1200/JCO.2022.40.16_suppl.8500 [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable – no new data is generated.