Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


The treatment of early-stage non-small cell lung cancer (NSCLC) is becoming increasingly complex. Standard of care management for the past decade has been adjuvant chemotherapy following curative intent resection regardless of nodal status or tumour profile. With the increased incorporation of immunotherapy in NSCLC, especially in the locally advanced, unresectable, or metastatic settings, multiple studies have sought to assess its utility in early-stage disease. While there are suboptimal responses to neoadjuvant chemotherapy alone, there is a strong rationale for the use of neoadjuvant immunotherapy in tumour downstaging, based upon the concept of enhanced T cell priming at the time of a high tumour antigen burden, and demonstrated clinically in other solid tumours, such as melanoma. In the NSCLC cancer setting, currently over 20 combinations of chemoimmunotherapy in the neoadjuvant and perioperative setting have been studied with results variable. Multiple large phase III studies have demonstrated that neoadjuvant chemoimmunotherapy combinations result in significant advances in pathological response, disease free and overall survival which has led to practice change across the world. Currently, combination immunotherapy regimens with novel agents targeting alternate immunomodulatory pathways are now being investigated. Given this, the landscape of treatment in resectable early-stage NSCLC has become increasingly complex. This review outlines the literature of neoadjuvant and perioperative immunotherapy and discusses its potential benefits and complexities and ongoing considerations into future research.

Free full text 


Logo of etatLink to Publisher's site
Explor Target Antitumor Ther. 2024; 5(6): 1247–1260.
Published online 2024 Sep 29. https://doi.org/10.37349/etat.2024.00273
PMCID: PMC11502072
PMID: 39465010

Evidence for the evolving role of neoadjuvant and perioperative immunotherapy in resectable non-small cell lung cancer

Thomas Hansen, Conceptualization, Investigation, Writing—original draft, Writing—review & editing, 1 Jonathon Hill, Validation, Writing—review & editing, Supervision, 2 , 3 , 4 Gary Tincknell, Validation, Writing—review & editing, Supervision, 2 , 3 , 4 , 5 Derrick Siu, Validation, Writing—review & editing, Supervision, 2 , 6 Daniel Brungs, Validation, Writing—review & editing, Supervision, 2 , 3 , 4 Philip Clingan, Validation, Writing—review & editing, Supervision, 2 , 3 , 4 Lorraine Chantrill, Validation, Writing—review & editing, Supervision, 2 , 4 and Udit Nindra, Conceptualization, Investigation, Writing—original draft, Writing—review & editing 2 , 7 , *
Stergios Boussios, Academic Editor and Nicola Normanno, Academic Editor
Stergios Boussios, King’s College London, UK;

Associated Data

Data Availability Statement

Abstract

The treatment of early-stage non-small cell lung cancer (NSCLC) is becoming increasingly complex. Standard of care management for the past decade has been adjuvant chemotherapy following curative intent resection regardless of nodal status or tumour profile. With the increased incorporation of immunotherapy in NSCLC, especially in the locally advanced, unresectable, or metastatic settings, multiple studies have sought to assess its utility in early-stage disease. While there are suboptimal responses to neoadjuvant chemotherapy alone, there is a strong rationale for the use of neoadjuvant immunotherapy in tumour downstaging, based upon the concept of enhanced T cell priming at the time of a high tumour antigen burden, and demonstrated clinically in other solid tumours, such as melanoma. In the NSCLC cancer setting, currently over 20 combinations of chemoimmunotherapy in the neoadjuvant and perioperative setting have been studied with results variable. Multiple large phase III studies have demonstrated that neoadjuvant chemoimmunotherapy combinations result in significant advances in pathological response, disease free and overall survival which has led to practice change across the world. Currently, combination immunotherapy regimens with novel agents targeting alternate immunomodulatory pathways are now being investigated. Given this, the landscape of treatment in resectable early-stage NSCLC has become increasingly complex. This review outlines the literature of neoadjuvant and perioperative immunotherapy and discusses its potential benefits and complexities and ongoing considerations into future research.

Keywords: Chemoimmunotherapy, resectable NSCLC, pathological complete response, overall survival, surgery

Introduction

Lung cancer is the leading cause of cancer-related mortality, accounting for approximately 1.8 million deaths annually [1]. Non-small cell lung cancer (NSCLC) is the commonest subtype, representing approximately 80% of lung cancers. In early-stage NSCLC, curative surgical resection remains the cornerstone of management. However, NSCLC is surgically resectable in about 25% of patients at time of diagnosis [2]. Currently, criteria for resectability in early-stage NSCLC vary by institution but generally include stage I to IIIA disease. Patients may have ipsilateral hilar nodal involvement but evidence of contralateral lung or nodal disease exclude patients from curative therapy. Furthermore, long-term outcomes following surgical resection, particularly for those with pathologically proven advanced nodal disease are poor, and the use of adjuvant chemotherapy only confers an approximate 5% improvement in 5-year overall survival (OS) [3].

In the locally advanced, unresectable, and metastatic settings, immunotherapy now plays a central role in standard of care management. This is driven by the improved outcomes seen in the PACIFIC study [4] with consolidation durvalumab in stage III unresectable patients, and the KEYNOTE-189 [5], CheckMate-227 [6] and IMPOWER-150 [7] studies in the metastatic setting. As a result, there has been a significant paradigm shift for the earlier incorporation of immunotherapy in NSCLC treatment. The theoretical benefits of a neoadjuvant IO approach in NSCLC are well documented. It is generally accepted that the presence of a full tumour mass upon commencement of immunotherapy enhances tumour immunogenicity through shedding of neoantigens at time of tumour cell death, thereby “priming” the T-cell tumour response as outlined in the schematic in Figure 1 [8]. Upfront resection of the tumour and associated tumour infiltrating lymphocytes (TILs) is therefore likely to negatively impact the intensity of the T-cell response that would be otherwise sustained from PD1/PD-L1 agents. Certainly, the augmented response of TIL activation using neoadjuvant treatment, compared to adjuvant therapy, is clearly demonstrated in pre-clinical breast cancer models [9]. Recently, this has been replicated in the clinical setting in resectable stage IIIB–IV melanoma, where neoadjuvant immunotherapy followed by surgery and adjuvant immunotherapy demonstrated greater clinical disease-free survival compared with surgery plus adjuvant immunotherapy alone [10, 11]. This was despite an identical duration of treatment in both arms. Currently pre-clinical models in lung and other solid cancer also support similar conclusions of improved TIL invasion into sites of disease and greater CD8 positive T-cell responses.

An external file that holds a picture, illustration, etc.
Object name is etat-05-1002273-g001.jpg

Neoadjuvant immune-checkpoint inhibitor activation in non-small cell lung cancer. Neoadjuvant immune checkpoint inhibitors (ICIs) can enhance T-cell priming through T-regulatory and immature dendritic cell pathways. Removal of the tumour during surgery is suspected to augment the response of tumour infiltrating lymphocytes, thereby potentially reducing the efficacy of ICIs. This forms the rationale for ongoing studies in the neoadjuvant lung cancer treatment pathway. Created in BioRender. Nindra, U. (2024) BioRender.com/w52g417

Given these strong rationales for the use of neoadjuvant immunotherapy there has been burgeoning interest in exploring its utility in the neoadjuvant space. CheckMate-816 was a landmark trial in this context, demonstrating that the addition of neoadjuvant nivolumab to chemotherapy resulted in significantly longer event-free survival and increased pathological complete response (pCR) compared to chemotherapy alone [12]. This study led to the first regulatory approval through the FDA of neoadjuvant chemoimmunotherapy in early non-small lung cancer, and paved the way for a multitude of further trials since investigating novel neoadjuvant immunotherapeutic approaches.

In light of this, the available evidence of immune-therapeutics in the neoadjuvant lung landscape is rapidly changing. We herein present a review of the currently reported evidence using neoadjuvant and perioperative approaches in resectable NSCLC. In addition, an insight into available predictive and prognostic biomarkers and the potential future directions of neoadjuvant immunotherapy in the precision medicine era. The following sections will highlight the current clinical trials that have been completed or are ongoing in the lung cancer space and their key results.

Our preliminary search identified 1,464 articles from which duplicates were removed and 1,166 were screened. 136 articles were retrieved for full review. Overall, 25 articles were included in the final analysis. We stratified the articles into four subgroups. This included neoadjuvant immunotherapy with chemotherapy, neoadjuvant immunotherapy plus radiotherapy, neoadjuvant immunotherapy alone, and perioperative chemo-immunotherapy. Data regarding treatment protocols, major pathological response (MPR), pCR, and survival data where available were extracted and are summarised in Table 1.

Table 1

Summary of studies investigating neoadjuvant immunotherapy in resectable lung cancer

Study Author, year Study phase Study protocol Study population Number of patients MPR (%) pCR (%) DFS/EFS/OS (%)
Trials investigating neoadjuvant immunotherapy + chemotherapy
NeoSTAR NCT03158129Cascone et al. [14], 2023IIArm 1: Nivolumab (x3)
Arm 2: Nivolumab + Ipilimumab (x1)
Arm 3: Nivolumab (x3) + CT
Arm 4: Nivolumab (x3) + Ipilimumab (x1) + CT
Resectable IA-IIIA NSCLCArm 1: 23
Arm 2: 21
Arm 3: 22
Arm 4: 22
22
38
32.1
50
9
29
18.2
18.2
12 m EFS: 96% (Arm 3) vs. 82% (Arm 4)
CheckMate-816 NCT02998528Spicer et al. [13], 2023IIIArm 1: Nivolumab + Platinum CT
Arm 2: Platinum CT
Resectable IB-IIIA NSCLCArm 1: 179
Arm 2: 179
36.9
8.9
24
2.2
31.6 m (95% CI, 30.2-NR) vs. 20.8 m (95% CI, 14.0 to 26.7) HR 0.63; P = 0.005).
24 m EFS: 63.8% vs. 45.3%
OS improvement of 13% in Nivolumab + CT group
No NCT IDNShen et al. [15], 2021IPembrolizumab 2 mg/kg + Nab-paclitaxel + Carboplatin (x2)Resectable stage IIB–IIIB squamous lung3745.964.9-
NCT04304248Zhao et al. [40], 2021 IIToripalimab + Carboplatin + Pemetrexed/Nab-paclitaxel (x3)Resectable stage IIIA or T3-4N2 IIIB NSCLC3360.645.5-
Lung-Mate001 ChiCTR1900023758Zhang et al. [37], 2021IISintilimab + Carboplatin + Pemetrexed/Gemcitabine) (x2-4)Resectable stage IB–IIIA5043.320-
Neo-Pre-IC NCT04326153Sun et al. [41], 2024 IISintilimab + Nab-paclitaxel + Carboplatin (x2-3)Resectable IIIA/IIIB NSCLC30654024 m DFS: 75%
No NCTDuan et al. [42], 2021ICT + ImmunotherapyStage IIA–IIIB NSCLC235030-
NCT02716038Shu et al. [43], 2020IIAtezolizumab + Carboplatin/Nab-paclitaxel (x2-4)Resectable IB-IIIA NSCLC305733-
NCT03480230Tfayli et al. [16], 2020IAvelumab (x4) + Cisplatin/Carboplatin + Gemcitabine/Pemetrexed (x3)Stage IB–IIIA NSCLC152211-
Trials investigating neoadjuvant immunotherapy
ChiCTR-OIC-17013726Gao et al. [44], 2020 IBSintilimab 200 mg (x2)Resectable stage IA–IIIB NSCLC4040.516.2-
CheckMate-159 NCT02259621Forde et al. [17], 2018; Reuss et al. [18], 2020IB/IIArm 1: Nivolumab (x2)
Arm 2: Nivolumab (x3) + Ipilimumab (x1)
Resectable IB-IIIA NSCLCArm 1: 21
Arm 2: 9
45
33.3
10
33.3
-
NEOMUN NCT03197467Eichhorn et al. [45], 2021IIPembrolizumab (x2)Resectable stage II–IIIA1527--
IONESCO NCT03030131Wislez et al. [20], 2022IIDurvalumab (x3)IB-IIIA (non-N2) resectable NSCLC4619312 m OS—89% (95% CI 75.8% to 95.3%)
12 m DFS—78% (95% CI 63.4% to 87.7%)
NeoCOAST NCT03794544Cascone et al. [19], 2023IIArm 1: Durvalumab
Arm 2: Durvalumab + Oleclumab
Arm 3: Durvalumab + Monalizumab
Arm 4: Durvalumab + Danvatirsen
Stage IA–IIIA NSCLCArm 1: 27
Arm 2: 21
Arm 3: 20
Arm 4: 16
12.5
22.2
33.3
33.3
3.7
9.5
10
12.5
-
NCT02927301Rusch et al. [46], 2023IIAtezolizumab (x2)Resectable stage IB–IIIB (non-EGFR/ALK)143206-
Trials investigating perioperative immunotherapy +/- chemotherapy
NADIM NCT03081689Provencio et al. [47], 2020IIPaclitaxel + Carboplatin + Nivolumab (x3) > Sx > NivolumabResectable stage IIIA NSCLC46836324 m PFS 77.1%
NADIM II NCT03838159Provencio et al. [25], 2022IIArm 1: Nivolumab + Platinum CT (x3) > Sx > Nivolumab (x6)
Arm 2: Platinum CT
Resectable stage IIIA–IIIB NSCLCArm 1: 57
Arm 2: 29
53
14
37
7
24 m OS 85% vs. 63.6% (HR 0.43)
24 m PFS 67.2% vs. 40.9% (HR 0.47)
TOP 1501 NCT02818920Tong et al. [48], 2022IIPembrolizumab (x2) > Sx > Pembrolizumab (x4)Stage IB–IIIA NSCLC3028--
KEYNOTE-671 NCT034256643Wakelee et al. [24], 2023IIIArm 1: Pembrolizumab + Platinum CT (x4) > Sx > Pembrolizumab (x13)
Arm 2: Placebo + Platinum CT (x4) > Sx > Placebo (x13)
Resectable stage II–IIIB NSCLC79730.2
11
18.1
4
24 m EFS—62.4% vs. 40.6% (HR 0.58)
24 m OS: 80.9% vs. 77.6% (P = 0.02)
NEOTORCH4 NCT04158440Lu et al. [49], 2024IIIArm 1: Toripalimab + Platinum CT (x3) > Sx > Toripalimab (x13)
Arm 2: Placebo + Platinum CT (x3) > Sx > Placebo (x13)
Resectable stage II–III NSCLCArm 1: 202
Arm 2: 202
48.5
8.4
24.8
1
mEFS: NE (95% CI, 24.4m–NE) vs. 15.1 m (95% CI, 10.6 m–21.9 m) (HR 0.40)
SAKK 16/14 NCT02572843Rothschild et al. [22], 2021IIDocetaxel + Cisplatin (x3) + Durvalumab (x2); > Sx > Durvalumab (x26)T1-3N2 stage IIIA NSCLC67621812 m EFS—73%
12 m OS—91%
AEGEAN NCT03800134Heymach et al. [23], 2023IIICT + Durvalumab or placebo (x3) > Sx > Durvalumab or placebo (x12)Resectable Stage II–IIIBArm 1: 366
Arm 2: 374
33.3
12.3
17.2
4.3
EFS NR (31.9 m–NR) vs. 25.9 m (18.9 m–NR), HR 0.68
NCT04316364Yan et al. [50], 2023IBAdebrelimab, Nab-paclitaxel + Carboplatin (x3) > Sx > Adebrelimab (x16)Resectable stage II–III NSCLC3751.429.7-
Trials investigating neoadjuvant IO with radiotherapy
NCT02904954Altorki et al. [51], 2021IIDurvalumab alone vs. Durvalumab + Stereotactic RTResectable stage I–IIIAArm 1: 30
Arm 2: 30
6.7
53.3
26-
NCT03030131 (IONESCO)Wislez et al. [20], 2022IIDurvalumab alone vs. Durvalumab + Stereotactic RTStage I–IIIAArm 1: 26
Arm 2: 26
---

m: month; NSCLC: non-small cell lung cancer; CT: chemotherapy; EFS: event free survival; NR: not reached; CI: confidence interval; NCT: National Clinical Trial; IDN: identification number; DFS: disease free survival; PFS: progression free survival; HR: hazard ratio; Sx: surgery; MPR: major pathological response; pCR: pathological complete response; OS: overall survival; EGFR: epidermal growth factor receptor; ALK: anaplastic lymphoma kina

Neoadjuvant immunotherapy and chemotherapy

In total, there were nine published trials investigating the effect of combination chemotherapy with immunotherapy involving six different immunotherapy agents. In respect to trials investigating nivolumab, CheckMate-816 is the largest phase III trial to date and evaluated the effectiveness of three cycles of neoadjuvant nivolumab with platinum-based chemotherapy compared to platinum-based chemotherapy alone in 358 randomised patients [12]. Chemoimmunotherapy was associated with a significantly longer event free survival [EFS; hazard ratio (HR) 0.63] in addition to a greater proportion of patients achieving pCR (24.0% vs. 2.2%). An exploratory analysis also revealed improved EFS in the subset of patients with pCR. Updated 3-year data has promoted the durability of this treatment response, with 3-year EFS of 57% compared to 43% (HR 0.68) in the nivolumab plus chemotherapy arm vs. chemotherapy alone groups respectively [13].

The phase II NeoSTAR trial expansion arms investigated the efficacy of neoadjuvant nivolumab and doublet nivolumab with ipilimumab with chemotherapy in patients with stage I to IIIA NSCLC [14]. In the 44 patients enrolled, the rate of MPR was 32.1% in the nivolumab plus chemotherapy group and 50% in the triplet cohort. When patients with known epidermal growth factor receptor (EGFR)/anaplastic lymphoma kina (ALK) alterations were excluded, the MPR rates improved further to 41.2% and 62.5%, respectively. The addition of ipilimumab appeared to reduce the extent of residual viable tumour, an effect which was more pronounced in the subset of patients with stage IIIA disease. This suggests that a subset of patients with more advanced nodal disease may benefit from dual checkpoint inhibition and merits further investigation in larger phase III studies. Importantly, the incorporation of CTLA-4 blockade was well tolerated without significant toxicities compared to anti-PD1 monotherapy.

Chemoimmunotherapy with pembrolizumab has also been assessed in the neoadjuvant NSCLC space. In a phase I study, Shen et al. [15] assessed the efficacy of neoadjuvant pembrolizumab with doublet chemotherapy in patients with stage IIB–IIIB squamous cell lung cancer, with pCR achieved in 17 of 37 (46%) patients. The only study which failed to demonstrate a benefit of this group was conducted by Tfayli et al. [16] in 2020. In a phase I cohort of 15 patients with stage IB–IIIA predominantly adenocarcinoma NSCLC, the addition of avelumab to chemotherapy did not result in an improved overall response rate compared to historical controls. Whether this negative result is a reflection of pharmacodynamic differences in avelumab compared with other PD1/PD-L1 therapies, or due to the small cohort size is unknown.

Neoadjuvant immunotherapy

Although most studies focus on chemoimmunotherapy as the preferred neoadjuvant approach to maximise response, there is data to demonstrate efficacy of immunotherapy as a monotherapy. The first trial data using neoadjuvant immunotherapy alone in resectable NSCLC emerged with initial results from Forde et al. [17] in 2018. A total of 20 patients completing 2 cycles of neoadjuvant nivolumab underwent radical surgery. There were no delays to surgery, no new safety signals, and a 45% MPR rate, with 90% achieving stable disease [17]. In light of these promising results, the same investigators opened a trial arm utilising neoadjuvant nivolumab with ipilimumab. Although this protocol demonstrated surgical feasibility, the study arm was discontinued after only 9 patients were enrolled due to toxicity, with 3 patients experiencing a grade ≥ 3 treatment related adverse events [18].

Arms 1 and 2 of NeoSTAR explored the efficacy of three doses of nivolumab, with arm 2 adding a single dose of ipilimumab in stage I–IIIA NSCLC [14]. 39 of 44 patients underwent curative surgery. The nivolumab and ipilimumab arm met its prespecified primary endpoint target of six MPRs, achieving this in 38% of patients (8/21). Furthermore, of those patients resected on trial, there was a higher pCR rate in the doublet arm (29% vs. 9%), without significant increase in toxicities. This highlights a promising role for combination checkpoint blockade in the neoadjuvant setting which may have an important place for those patients who are not suitable candidates for systemic chemotherapy. The benefit of combination therapy was also suggested in the NEOCOAST study which investigated durvalumab alone or in combination with other immune modulating therapies [19]. In patients who received neoadjuvant durvalumab alone, MPR rates were 11% but those who were treated with concomitant oleculumab (anti-CD73), monalizumab (anti-NKG2A) or danvatisern (anti-STAT3-antisense-oligonucleotide) had MPR rates of 19%, 30% and 31%, respectively. These collectively encouraging results support the need for further evaluation of combination immunotherapy in the neoadjuvant NSCLC setting.

More recently the IONESCO trial assessed the feasibility of three cycles of neoadjuvant durvalumab in patients with resectable NSCLC. Of the 43 patients proceeding to surgery, 8 (19%) achieved MPR, with 12-month OS and disease free survival (DFS) rates 89% and 78% respectively [20]. NEOMUN was a further phase II single-arm study which administered two cycles of pembrolizumab prior to surgery, with a comparable 27% MPR [21].

Perioperative immunotherapy +/- chemotherapy

Perioperative immunotherapy, with and without chemotherapy, has been investigated in 7 recent trials. Of these, SAKK 14/16 was the earliest to be reported, assessing the utility of chemotherapy with cisplatin and docetaxel with sequential durvalumab using two cycles pre-operatively and 12 months in the adjuvant setting for stage IIIA NSCLC with N2 disease. This study yielded R0 resection rates of 93% (51/55, 93%) and a 73% 1-year EFS [22]. Following on from this, the efficacy of peri-operative durvalumab was reported by the AEGEAN investigators in a large phase III randomised trial comparing 12 cycles of post-operative durvalumab to placebo following 3 cycles of neoadjuvant chemo-immunotherapy in patients with stage II–IIIB resectable NSCLC [23]. The primary endpoints of this trials were EFS and PCR, in a modified intention to treat subset of 740 patients without actionable EGFR or ALK alterations. EFS was demonstrated in 73.4% of patients receiving durvalumab compared to 64.5% in the placebo group with a HR of 0.68, in addition to 17.2% vs. 4.3% pCR respectively [13% difference; 95% confidence interval (CI) 8.7 to 17.6; P < 0.001].

These results were very comparable to the largest of the perioperative trials, the phase III KEYNOTE-671 [24]. This randomized patients into perioperative pembrolizumab with chemotherapy vs. neoadjuvant chemotherapy alone. pCR rate was 18.1% in this study and 24-month EFS rates were 62.4% in the pembrolizumab group vs. 40.6% in the placebo arm (HR 0.58), although not yet enough to demonstrate a clear OS benefit at 2-years (80.9% vs. 77.6%). Interestingly, the highest pCR rates were achieved in the NADIM studies despite enrolling only stage IIIA patients. In the phase II NADIM trial, patients received 3 cycles of neoadjuvant carboplatin, paclitaxel and nivolumab, followed by surgery, then 12 months of adjuvant nivolumab. Of the 41 patients undergoing surgery, 34 (83%; 95% CI 68–93) had MPR, with 26 (63%) achieving pCR. The 3-year OS was 81.9% in the intention-to-treat population (Provencio et al. [25]). The NADIM II trial randomised patients to this same neoadjuvant chemoimmunotherapy regimen and compared it to chemotherapy alone. They also reported an excellent 37% pCR rate (although inferior to NADIM) in the combination group vs. 7% in the comparator arm (RR 5.34; 95% CI 1.34 to 21.23; P = 0.02) [25]. The only reason found to potentially account for the vast difference in pCR rates in the two NADIM trials was the higher carboplatin dose used in the latter (AUC6 vs. AUC5). The reasons for their superiority over CheckMate-816 [26] are interesting, though as yet unexplained.

Summation

Overall, the results of the above trials support the clear role of neoadjuvant immunotherapy in early NSCLC. There is a distinct and demonstrable effect on enhancing pCR and MPR that is consistently observed in its use as monotherapy and in combination with chemotherapy, and extending into the perioperative setting. Furthermore, it offers higher rates of R0 resection without surgical delays, and does this with a tolerable side-effect profile. However, and perhaps of little surprise given the tempo of evolution in the field, there is significant heterogeneity across these trials in the selection of checkpoint inhibitor, the duration of their use pre- and post-operatively, and clinical endpoints measured.

Safety of neoadjuvant immunotherapy

With additional agents being used in the cancer paradigm, especially in the curative setting, there is always concern about increasing toxicity. Recently, a meta-analysis of sixteen studies across multiple tumour streams demonstrated that grade ≥ 3 immune related adverse events was 24.0% with led to discontinuation of neoadjuvant protocols in 9.4% of patients [27]. In the NSCLC setting, a recent systematic review has shown no difference in grade 3–5 treatment related adverse events between patients receiving neoadjuvant chemoimmunotherapy vs. those receiving neoadjuvant chemotherapy only [28]. In fact, patients who had chemoimmunotherapy were more likely to have surgery and less likely to have progression of disease that prevented curative intent surgery. No differences in patient refusal for surgery has been demonstrated between chemoimmunotherapy and chemotherapy populations [28].

Pathological responses as surrogate markers of survival

pCR is a key primary endpoint in a number of neoadjuvant studies and is defined as 0% residual viable tumour. Currently, the role of pCR as a confirmed surrogate endpoint of either recurrence free survival or OS is not well established. MPR defined arbitrarily as less than 10% residual viable tumour is an alternatively used measure [29]. Studies in the neoadjuvant chemotherapy and chemo-radiotherapy resectable NSCLC settings have supported the role of pCR as a predictive marker of OS [30, 31]. In 2020, a systematic review of neoadjuvant therapy of any kind in resectable lung cancer also supported the role of PCR as a surrogate marker of OS using 33 studies. They demonstrated that for OS by PCR status, the HRs ranged from 0.13 to 0.78, and the meta-analysed HR across 21 studies (6,672 patients) was 0.49 (95% CI 0.43–0.56) [32]. Recently, authors from the CheckMate-816 study investigated the influence of residual tumour volume (RVT) on EFS as defined by either recurrence or death by any cause [33]. They demonstrated that RVT was predictive of EFS with 2-year EFS rates of 90%, 60%, 57%, and 39% for patients with 0–5%, 5–30%, 30–80%, and > 80% RVT. This relationship was sustained despite the presence or absence of lymph node involvement of the original tumour. These results are the most recent in the neoadjuvant immunotherapy space and greatly support the use of PCR and RVT as appropriate surrogate markers of long-term progression free and OS.

Predictive markers of immunotherapy response

The use of predictive molecular markers to identify patients with higher likelihood of response is of paramount importance and a number of these have been recognised in the literature and summarised in Figure 2. PD-L1 expression is recognised as an important indicator of immune-responsiveness in the metastatic setting [5]. Follow-up analysis of the LCMC3 cohort revealed a strong correlation between PD-L1 TPS and major pathological response [34]. The NEOMUN trial echoed these results, demonstrating an augmented pathologic response with pembrolizumab in patients with higher PD-L1 [21]. CheckMate-816 demonstrated that although benefit of nivolumab with chemotherapy was observed across all subgroups, the benefit was most pronounced in patients with PD-L1 expression of > 1%, although the trial was not statistically powered for this analysis [12]. A meta-analysis of neoadjuvant chemoimmunotherapy and chemotherapy in NSCLC similarly demonstrated benefit in EFS across all PD-L1 subgroups for chemoimmunotherapy [28]. However, a consistent theme observed across all of the above trials was that pCR is seen in patients with PD-L1 ≤ 1%. Although this might be explained by intratumoral heterogeneity, it highlights the need for further evaluation of the predictive ability of differential PD-L1 levels in prospective PD-L1 all-comer trials. In addition to PD-L1, tumour mutational burden (TMB) has also been shown to be a potential predictive biomarker for neoadjuvant immunotherapy in NSCLC [17]. Interestingly, in the same study, Forde et al. [17] did not note a correlation between TMB and PD-L1 expression thereby highlighting that these two biomarkers may act as independent predictive tools for clinicians in future. TMB has much more evidence in the metastatic NSCLC as being a predictive marker of immunotherapy response [35] and based on the KEYNOTE-158 study, the FDA has approved blanket use of pembrolizumab in patients with advanced cancer with high TMB [36].

An external file that holds a picture, illustration, etc.
Object name is etat-05-1002273-g002.jpg

Predictive markers of immunotherapy in non-small cell lung cancer. Multiple predictive markers of immunotherapy have been investigated over the past few decades. Currently tumour mutational burden (TMB) and PD-L1 expression remain the two most clinically used markers but increasing data regarding the role of inflammation, gut microbiome and tumour microenvironment are emerging. Further, presence of oncogenic driver mutations such as EGFR and ALK also appear to dampen the likelihood of immunotherapy response in advanced NSCLC but their role as predictive markers in the neoadjuvant setting are currently unknown. ALK: anaplastic lymphoma kina. Created in BioRender. Nindra, U. (2024) BioRender.com/k45y858

The use of circulating tumour DNA (ctDNA) is becoming increasingly recognised as a promising surrogate marker for minimal residual disease and is gaining interest in the immunotherapy setting. The NADIM II trial demonstrated that low pre-treatment and post-treatment ctDNA levels were associated with significantly improved progression-free survival and OS [25]. An exploratory analysis of pre-operative ctDNA was also assessed in the CheckMate-816 cohort, which demonstrated a higher percentage of ctDNA clearance with nivolumab plus chemotherapy (56%; 95% CI 40–71) than with chemotherapy alone (35%; 95% CI 21–51), with a higher EFS in those patients achieving ctDNA clearance across both treatment arms [12]. Longitudinal assessment of ctDNA following resection of operable NSCLC has revealed that detectable ctDNA in the post-operative setting is associated with early recurrence and poor prognosis [37]. Moreover, the relapse risk in ctDNA negative patients did not change with receipt of adjuvant chemotherapy, heralding its potential utility as a tool to select patients suitable for adjuvant immunotherapy de-escalation in the future. This however requires further investigation of its use in dedicated perioperative immunotherapy trials.

There has also been increasing research into the role of gut microbiota and response to immune checkpoint inhibitors. Recently, it was shown that elevated diversity and composition of microbiota was correlated with improved response to immunotherapy [38]. Part of this response was thought to be due to presence of faecalibacterium that was accompanied by increased presence of short-chain fatty acids. Additionally, alterations in gut microbiota through the use of probiotics has also been correlated with improved immunotherapy response in NSCLC [39]. Whether this is a direct result of probiotic use or the result of changes in gut flora requires ongoing research. Given the suggestion of gut microbiota diversity, faecal transplantation has been postulated to augment immunotherapy response with some evidence in animal models of improved outcomes [38, 39]. However, further research in this space is required before human trials are explored.

Conclusion

Overall, it is evident that based on multiple studies that concomitant neoadjuvant immunotherapy in addition to chemotherapy provides PCR and potential OS benefit. There is still an unmet need to identify patients who are most likely to benefit from therapy, as well as the ideal combination of immunotherapy regimen in the neoadjuvant setting. In addition to this, for those who do not achieve a PCR and therefore identified as high risk of recurrent disease, the optimal adjuvant treatment, including treatment intensification, remains unclear. Overall, the field of early lung cancer treatment is expanding and becoming increasingly complex and with neoadjuvant immunotherapy now becoming mainstay in other tumour streams such as breast cancer, increasing capacity to determine which patients would most benefit is essential. Future directions will need to be focused not only standardised treatment guidelines but also predictive clinical, biological, and potentially radiological markers of response and survival.

Abbreviations

ALKanaplastic lymphoma kina
CIconfidence interval
ctDNAcirculating tumour DNA
DFSdisease free survival
EFSevent free survival
EGFRepidermal growth factor receptor
HRhazard ratio
MPRmajor pathological response
NSCLCnon-small cell lung cancer
OSoverall survival
pCRpathological complete response
RVTresidual tumour volume
TMBtumour mutational burden

Contributor Information

Stergios Boussios, King’s College London, UK.

Nicola Normanno, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Italy.

Declarations

Author contributions

TH: Conceptualization, Investigation, Writing—original draft, Writing—review & editing. JH: Validation, Writing—review & editing, Supervision. GT: Validation, Writing—review & editing, Supervision. DS: Validation, Writing—review & editing, Supervision. DB: Validation, Writing—review & editing, Supervision. PC: Validation, Writing—review & editing, Supervision. LC: Validation, Writing—review & editing, Supervision. UN: Conceptualization, Investigation, Writing—original draft, Writing—review & editing. All authors read and approved the submitted version.

Conflicts of interest

The authors declare that they have no conflicts of interest.

Ethical approval

Not applicable.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of data and materials

Both Figures 1 and 2 were created using BioRender, please refer to the relevant section in the main text for specific information.

Funding

Not applicable.

Copyright

© The Author(s) 2024.

References

1. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet. 2021;398:535–54. 10.1016/S0140-6736(21)00312-3. [Abstract] [CrossRef] [Google Scholar]
2. Thandra KC, Barsouk A, Saginala K, Aluru JS, Barsouk A. Epidemiology of lung cancer. Contemp Oncol (Pozn) 2021;25:45–52. 10.5114/wo.2021.103829. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
3. NSCLC Meta-analysis Collaborative Group Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet. 2014;383:1561–71. 10.1016/S0140-6736(13)62159-5. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-Year Survival Outcomes From the PACIFIC Trial: Durvalumab After Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. J Clin Oncol. 2022;40:1301–11. 10.1200/JCO.21.01308. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. KEYNOTE-189 Investigators Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med. 2018;378:2078–92. 10.1056/NEJMoa1801005. [Abstract] [CrossRef] [Google Scholar]
6. Borghaei H, O’Byrne KJ, Paz-Ares L, Ciuleanu TE, Yu X, Pluzanski A, et al. Nivolumab plus chemotherapy in first-line metastatic non-small-cell lung cancer: results of the phase III CheckMate 227 Part 2 trial. ESMO Open. 2023;8:102065. 10.1016/j.esmoop.2023.102065. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
7. Socinski MA, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, et al. IMpower150 Final Overall Survival Analyses for Atezolizumab Plus Bevacizumab and Chemotherapy in First-Line Metastatic Nonsquamous NSCLC. J Thorac Oncol. 2021;16:1909–24. 10.1016/j.jtho.2021.07.009. [Abstract] [CrossRef] [Google Scholar]
8. Blank CU, Rozeman EA, Fanchi LF, Sikorska K, van de Wiel B, Kvistborg P, et al. Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med. 2018;24:1655–61. 10.1038/s41591-018-0198-0. [Abstract] [CrossRef] [Google Scholar]
9. Liu J, Blake SJ, Yong MC, Harjunpää H, Ngiow SF, Takeda K, et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov. 2016;6:1382–99. 10.1158/2159-8290.CD-16-0577. [Abstract] [CrossRef] [Google Scholar]
10. Patel SP, Othus M, Chen Y, Wright GP Jr, Yost KJ, Hyngstrom JR, et al. Neoadjuvant-Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N Engl J Med. 2023;388:813–23. 10.1056/NEJMoa2211437. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
11. Blank CU, Lucas MW, Scolyer RA, van de Wiel BA, Menzies AM, Lopez-Yurda M, et al. Neoadjuvant Nivolumab and Ipilimumab in Resectable Stage III Melanoma. N Engl J Med. 2024;[Epub ahead of print] 10.1056/NEJMoa2402604. [Abstract] [CrossRef] [Google Scholar]
12. Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. CheckMate 816 Investigators Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med. 2022;386:1973–85. 10.1056/NEJMoa2202170. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Spicer J, Forde PM, Provencio M, Lu S, Wang C, Mitsudomi T, et al. Clinical outcomes with neoadjuvant nivolumab (N) + chemotherapy (C) vs C by definitive surgery in patients (pts) with resectable NSCLC: 3-y results from the phase 3 CheckMate 816 trial. J Clin Oncol. 2023;41:8521. 10.1200/JCO.2023.41.16_suppl.8521. [CrossRef] [Google Scholar]
14. Cascone T, Leung CH, Weissferdt A, Pataer A, Carter BW, Godoy MCB, et al. Neoadjuvant chemotherapy plus nivolumab with or without ipilimumab in operable non-small cell lung cancer: the phase 2 platform NEOSTAR trial. Nat Med. 2023;29:593–604. 10.1038/s41591-022-02189-0. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Shen D, Wang J, Wu J, Chen S, Li J, Liu J, et al. Neoadjuvant pembrolizumab with chemotherapy for the treatment of stage IIB-IIIB resectable lung squamous cell carcinoma. J Thorac Dis. 2021;13:1760–8. 10.21037/jtd-21-103. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Tfayli A, Al Assaad M, Fakhri G, Akel R, Atwi H, Ghanem H, et al. Neoadjuvant chemotherapy and Avelumab in early stage resectable nonsmall cell lung cancer. Cancer Med. 2020;9:8406–11. 10.1002/cam4.3456. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. 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;378:1976–86. 10.1056/NEJMoa1716078. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Reuss JE, Anagnostou V, Cottrell TR, Smith KN, Verde F, Zahurak M, et al. Neoadjuvant nivolumab plus ipilimumab in resectable non-small cell lung cancer. J Immunother Cancer. 2020;8:e001282. 10.1136/jitc-2020-001282. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Cascone T, Kar G, Spicer JD, García-Campelo R, Weder W, Daniel DB, et al. Neoadjuvant Durvalumab Alone or Combined with Novel Immuno-Oncology Agents in Resectable Lung Cancer: The Phase II NeoCOAST Platform Trial. Cancer Discov. 2023;13:2394–411. 10.1158/2159-8290.CD-23-0436. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
20. Wislez M, Mazieres J, Lavole A, Zalcman G, Carre O, Egenod T, et al. Neoadjuvant durvalumab for resectable non-small-cell lung cancer (NSCLC): results from a multicenter study (IFCT-1601 IONESCO) J Immunother Cancer. 2022;10:e005636. 10.1136/jitc-2022-005636. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Eichhorn F, Klotz LV, Bischoff H, Thomas M, Lasitschka F, Winter H, et al. Neoadjuvant anti-programmed Death-1 immunotherapy by Pembrolizumab in resectable nodal positive stage II/IIIa non-small-cell lung cancer (NSCLC): the NEOMUN trial. BMC Cancer. 2019;19:413. 10.1186/s12885-019-5624-2. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
22. Rothschild SI, Zippelius A, Eboulet EI, Savic Prince S, Betticher D, Bettini A, et al. Swiss Group for Clinical Cancer Research (SAKK) 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;39:2872–80. 10.1200/JCO.21.00276. [Abstract] [CrossRef] [Google Scholar]
23. Heymach JV, Harpole D, Mitsudomi T, Taube JM, Galffy G, Hochmair M, et al. AEGEAN Investigators Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer. N Engl J Med. 2023;389:1672–84. 10.1056/NEJMoa2304875. [Abstract] [CrossRef] [Google Scholar]
24. Wakelee H, Liberman M, Kato T, Tsuboi M, Lee SH, Gao S, et al. KEYNOTE-671 Investigators Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer. N Engl J Med. 2023;389:491–503. 10.1056/NEJMoa2302983. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
25. 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;40:2924–33. 10.1200/JCO.21.02660. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Spicer J, Girard N, Provencio M, Wang C, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab (NIVO) + chemotherapy (chemo) vs chemo in patients (pts) with resectable NSCLC: 4-year update from CheckMate 816. J Clin Oncol. 2024;42:LBA8010. 10.1200/JCO.2024.42.17_suppl.LBA8010. [CrossRef] [Google Scholar]
27. Feng Y, Guo K, Jin H, Jiang J, Wang M, Lin S. Adverse events of neoadjuvant combination immunotherapy for resectable cancer patients: a systematic review and meta-analysis. Front Immunol. 2024;14:1269067. 10.3389/fimmu.2023.1269067. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
28. Sorin M, Prosty C, Ghaleb L, Nie K, Katergi K, Shahzad MH, et al. Neoadjuvant Chemoimmunotherapy for NSCLC: A Systematic Review and Meta-Analysis. JAMA Oncol. 2024;10:621–33. 10.1001/jamaoncol.2024.0057. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Saqi A, Leslie KO, Moreira AL, Lantuejoul S, Shu CA, Rizvi NA, et al. Assessing Pathologic Response in Resected Lung Cancers: Current Standards, Proposal for a Novel Pathologic Response Calculator Tool, and Challenges in Practice. JTO Clin Res Rep. 2022;3:100310. 10.1016/j.jtocrr.2022.100310. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Mouillet G, Monnet E, Milleron B, Puyraveau M, Quoix E, David P, et al. Intergroupe Francophone de Cancérologie Thoracique (IFCT) Pathologic complete response to preoperative chemotherapy predicts cure in early-stage non-small-cell lung cancer: combined analysis of two IFCT randomized trials. J Thorac Oncol. 2012;7:841–9. 10.1097/JTO.0b013e31824c7d92. [Abstract] [CrossRef] [Google Scholar]
31. Schreiner W, Gavrychenkova S, Dudek W, Rieker RJ, Lettmaier S, Fietkau R, et al. Pathologic complete response after induction therapy-the role of surgery in stage IIIA/B locally advanced non-small cell lung cancer. J Thorac Dis. 2018;10:2795–803. 10.21037/jtd.2018.05.68. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
32. 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. [CrossRef] [Google Scholar]
33. Deutsch JS, Cimino-Mathews A, Thompson E, Provencio M, Forde PM, Spicer J, et al. Association between pathologic response and survival after neoadjuvant therapy in lung cancer. Nat Med. 2024;30:218–28. 10.1038/s41591-023-02660-6. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
34. Chaft JE, Shyr Y, Sepesi B, Forde PM. Preoperative and Postoperative Systemic Therapy for Operable Non-Small-Cell Lung Cancer. J Clin Oncol. 2022;40:546–55. 10.1200/JCO.21.01589. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Kalvapudi S, Vedire Y, Yendamuri S, Barbi J. Neoadjuvant therapy in non-small cell lung cancer: basis, promise, and challenges. Front Oncol. 2023;13:1286104. 10.3389/fonc.2023.1286104. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
36. Maio M, Ascierto PA, Manzyuk L, Motola-Kuba D, Penel N, Cassier PA, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol. 2022;33:929–38. 10.1016/j.annonc.2022.05.519. [Abstract] [CrossRef] [Google Scholar]
37. Qiu B, Guo W, Zhang F, Lv F, Ji Y, Peng Y, et al. Dynamic recurrence risk and adjuvant chemotherapy benefit prediction by ctDNA in resected NSCLC. Nat Commun. 2021;12:6770. 10.1038/s41467-021-27022-z. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
38. Ren S, Feng L, Liu H, Mao Y, Yu Z. Gut microbiome affects the response to immunotherapy in non-small cell lung cancer. Thorac Cancer. 2024;15:1149–63. 10.1111/1759-7714.15303. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
39. Del Giudice T, Staropoli N, Tassone P, Tagliaferri P, Barbieri V. Gut Microbiota Are a Novel Source of Biomarkers for Immunotherapy in Non-Small-Cell Lung Cancer (NSCLC) Cancers (Basel) 2024;16:1806. 10.3390/cancers16101806. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
40. Zhao ZR, Yang CP, Chen S, Yu H, Lin YB, Lin YB, et al. Phase 2 trial of neoadjuvant toripalimab with chemotherapy for resectable stage III non-small-cell lung cancer. Oncoimmunology. 2021;10:1996000. 10.1080/2162402X.2021.1996000. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Sun C, Wang X, Xu Y, Shao G, Chen X, Liu Y, et al. Efficiency and safety of neoadjuvant PD-1 inhibitor (sintilimab) combined with chemotherapy in potentially resectable stage IIIA/IIIB non-small cell lung cancer: Neo-Pre-IC, a single-arm phase 2 trial. EClinicalMedicine. 2024;68:102422. 10.1016/j.eclinm.2024.102422. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
42. Duan H, Wang T, Luo Z, Tong L, Dong X, Zhang Y, et al. Neoadjuvant programmed cell death protein 1 inhibitors combined with chemotherapy in resectable non-small cell lung cancer: an open-label, multicenter, single-arm study. Transl Lung Cancer Res. 2021;10:1020–8. 10.21037/tlcr-21-130. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
43. Shu CA, Gainor JF, Awad MM, Chiuzan C, Grigg CM, Pabani A, et al. Neoadjuvant atezolizumab and chemotherapy in patients with resectable non-small-cell lung cancer: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21:786–95. 10.1016/S1470-2045(20)30140-6. [Abstract] [CrossRef] [Google Scholar]
44. 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;15:816–26. 10.1016/j.jtho.2020.01.017. [Abstract] [CrossRef] [Google Scholar]
45. Eichhorn F, Klotz LV, Kriegsmann M, Bischoff H, Schneider MA, Muley T, et al. Neoadjuvant anti-programmed death-1 immunotherapy by pembrolizumab in resectable non-small cell lung cancer: First clinical experience. Lung Cancer. 2021;153:150–7. 10.1016/j.lungcan.2021.01.018. [Abstract] [CrossRef] [Google Scholar]
46. Rusch VW, Nicholas A, Patterson GA, Waqar SN, Toloza EM, Haura EB, et al. Surgical results of the Lung Cancer Mutation Consortium 3 trial: A phase II multicenter single-arm study to investigate the efficacy and safety of atezolizumab as neoadjuvant therapy in patients with stages IB-select IIIB resectable non-small cell lung cancer. J Thorac Cardiovasc Surg. 2023;165:828–39.e5. 10.1016/j.jtcvs.2022.10.007. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
47. 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;21:1413–22. 10.1016/S1470-2045(20)30453-8. [Abstract] [CrossRef] [Google Scholar]
48. Tong BC, Gu L, Wang X, Wigle DA, Phillips JD, Harpole DH Jr, et al. Perioperative outcomes of pulmonary resection after neoadjuvant pembrolizumab in patients with non-small cell lung cancer. J Thorac Cardiovasc Surg. 2022;163:427–36. 10.1016/j.jtcvs.2021.02.099. [Abstract] [CrossRef] [Google Scholar]
49. Lu S, Zhang W, Wu L, Wang W, Zhang P, Neotorch Investigators. Fang W, Xing W, Chen Q, Yang L, Mei J, Tan L, et al. Perioperative Toripalimab Plus Chemotherapy for Patients With Resectable Non-Small Cell Lung Cancer: The Neotorch Randomized Clinical Trial. JAMA. 2024;331:201–11. 10.1001/jama.2023.24735. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
50. Yan W, Zhong WZ, Liu YH, Chen Q, Xing W, Zhang Q, 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. 2023;18:194–203. 10.1016/j.jtho.2022.09.222. [Abstract] [CrossRef] [Google Scholar]
51. Altorki NK, McGraw TE, Borczuk AC, Saxena A, Port JL, Stiles BM, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021;22:824–35. 10.1016/S1470-2045(21)00149-2. [Abstract] [CrossRef] [Google Scholar]

Articles from Exploration of Targeted Anti-tumor Therapy are provided here courtesy of Open Exploration Publishing

Citations & impact 


This article has not been cited yet.

Impact metrics

Alternative metrics

Altmetric item for https://www.altmetric.com/details/169076032
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/169076032

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.