Advancing neoadjuvant immunotherapy in non-small cell lung cancer (NSCLC): commentary on the Lung Cancer Mutation Consortium 3 trial (LCMC3)
Editorial Commentary

Advancing neoadjuvant immunotherapy in non-small cell lung cancer (NSCLC): commentary on the Lung Cancer Mutation Consortium 3 trial (LCMC3)

Lanyi N. Chen1 ORCID logo, Bryan P. Stanifer2, Joshua R. Sonett2, Catherine A. Shu1

1Division of Hematology and Oncology, Columbia University Medical Center, New York, NY, USA; 2Department of Surgery, Columbia University Medical Center, New York, NY, USA

Correspondence to: Catherine A. Shu, MD. Division of Hematology and Oncology, Columbia University Medical Center, 161 Fort Washington Ave, 3rd Floor, New York, NY 10032, USA. Email: cas2145@cumc.columbia.edu.

Comment on: Rusch VW, Nicholas A, Patterson GA, 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 nonsmall cell lung cancer. J Thorac Cardiovasc Surg 2023;165:828-39.e5.


Keywords: Neoadjuvant; chemoimmunotherapy; non-small cell lung cancer (NSCLC); surgical outcomes


Received: 13 February 2024; Accepted: 03 June 2024; Published online: 21 June 2024.

doi: 10.21037/actr-24-13


Introduction

The perioperative management of resectable non-small cell lung cancer (NSCLC) has evolved from adjuvant systemic therapy to an added focus on neoadjuvant therapy, which is thought to have increased efficacy due to the presence of intact tumor and ability to induce tumor shrinkage prior to surgery (1). Findings from the Lung Cancer Mutation Consortium 3 (LCMC3) trial, the largest neoadjuvant immunotherapy trial, established a benefit with checkpoint inhibitor monotherapy and set the stage for subsequent trials studying neoadjuvant and perioperative chemoimmunotherapy (2). The landmark Checkmate 816 and KEYNOTE-671 trials, for instance, led to recent Food and Drug Administration (FDA) approvals for neoadjuvant and perioperative immunotherapy in combination with chemotherapy, respectively (3,4). However, several questions remain regarding the optimal perioperative management in this diverse patient population, including the role of biomarkers in treatment selection, the identification of patients who may benefit from immunotherapy alone rather than combined chemoimmunotherapy, and the impact of perioperative therapies on surgical approach and outcomes (5,6).

A recent report on pathologic and surgical outcomes in patients enrolled in LCMC3, which confirmed the benefit of neoadjuvant immunotherapy, provides important contributions to ongoing discussion regarding patient selection and multidisciplinary care within this rapidly changing therapeutic context (7). Here, we review the findings reported from LCMC3 and interpret them in the context of more recently published randomized chemoimmunotherapy trials. We reflect on how the results of LCMC3 provide a foundation upon which to further discuss patient selection for perioperative treatments and optimization of the multidisciplinary treatment approach for resectable NSCLC.


LCMC3 overview and summary: pathologic and surgical outcomes

LCMC3 was a multicenter, single-arm phase II study that investigated the programmed death ligand 1 (PD-L1) checkpoint inhibitor atezolizumab as neoadjuvant treatment for patients with untreated, resectable stage IB–IIIB NSCLC (7). A total of 181 patients were enrolled and treated with 2 cycles of atezolizumab prior to surgical resection, followed by up to 12 months of adjuvant atezolizumab in patients with evidence of pathologic response or lack of radiographic progression. The primary endpoint of the study was major pathological response (MPR), defined as 10% or less viable tumor tissue at the time of surgical resection. Secondary endpoints included pathologic complete response (pCR) and radiographic response (7).

Most (81%) patients had stage IIB–IIIB tumors: 18 (10%) had stage IB disease, 16 (9%) had IIA disease, 55 (30%) had IIB disease, 70 (39%) had IIIA disease, and 22 (12%) had IIIB disease. Most patients (90%) were current or former smokers, and 38% of tumors had squamous histology. The trial population was unselected for PD-L1; 27% of tumors had a tumor proportion score (TPS) for PD-L1 of ≥50% and 15% had a TPS of 1–49% (7). All 181 patients received at least 1 cycle of neoadjuvant atezolizumab, and 88% (n=159) proceeded to surgery. Of the 22 patients (12%) who did not undergo surgery, 10 (6%) had evidence of radiographic progression, 6 (3%) did not proceed with surgery due to physician decision, 3 (2%) withdrew consent, and 3 (2%) were noted to have “other” reasons: 1 patient was noted to have congestive heart failure, 1 declined surgery, and 1 was lost to follow-up.

The primary efficacy population (n=143) that was used for analysis included all patients who underwent surgery, but excluded 16 patients with EGFR mutations or ALK translocations due to emerging evidence that the benefit of immunotherapy is limited in this patient population (8,9). Within the primary efficacy population, an MPR was observed in 20% (n=29 of 143) of patients, and pCR was observed in 6% (n=8 of 143), as noted in Table 1 (3,7,10-17). Of note, while evaluation of pCR is generally thought to be more reliable, interobserver reproducibility has been demonstrated for MPR assessment (18). In line with findings from other neoadjuvant trials, there was a lack of association between radiographic response and final pathological stage in LCMC3. This highlights the importance of the latter in accurately assessing response to neoadjuvant immunotherapy.

Table 1

Pathologic outcomes in LCMC3 and other neoadjuvant and perioperative trials

Trial Intervention Patient population Rate of MPR Rate of PCR
LCMC3 (7) Neoadjuvant atezolizumab 1,200 mg (2 cycles q3 weeks) and adjuvant atezolizumab (up to 12 months) Stage IB–IIIB NSCLC, n=181 (n=143 in efficacy population) 20% 6%
NEOSTAR (10,11) Neoadjuvant nivolumab 3 mg/kg (3 cycles q2 weeks) vs. neoadjuvant nivolumab 3 mg/kg plus ipilimumab 1 mg/kg (3 cycles q2 weeks) Stage I–IIIA NSCLC, n=44 (n=23 to nivolumab; n=21 to nivolumab + ipilimumab) Nivolumab: 22% Nivolumab: 9%
Nivolumab + ipilimumab: 38% Nivolumab + ipilimumab: 29%
KEYNOTE-671 (3) Neoadjuvant pembrolizumab 200 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant pembrolizumab (q3 weeks for up to 13 cycles) vs. neoadjuvant chemotherapy plus placebo (q3 weeks for 4 cycles) and adjuvant placebo (q3 weeks for up to 13 cycles) Stage II–IIIB NSCLC, n=797 (n=397 to pembrolizumab; n=400 to placebo) Pembrolizumab + chemotherapy: 30.2% Pembrolizumab + chemotherapy: 18.1%
Placebo + chemotherapy: 11.0% Placebo + chemotherapy: 4.0%
Checkmate-816 (12) Neoadjuvant nivolumab 360 mg plus platinum-based chemotherapy (q3 weeks for 3 cycles) vs. neoadjuvant platinum-based chemotherapy (q3 weeks for 3 cycles) Stage IB–IIIA NSCLC; n=358 (n=179 to nivolumab; n=179 to chemotherapy only) Nivolumab + chemotherapy: 37% Nivolumab + chemotherapy: 24%
Chemotherapy only: 8.9% Chemotherapy only: 2.2%
AEGEAN (13) Neoadjuvant durvalumab 1,500 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant durvalumab (q4 weeks for up to 12 cycles) vs. neoadjuvant platinum-based chemotherapy plus placebo (q3 weeks for 4 cycles) and adjuvant placebo (q4 weeks for up to 12 cycles) Stage IIA to IIIB NSCLC; n=802 (n=400 to durvalumab; n=402 to placebo) Durvalumab + chemotherapy: 33.3% Durvalumab + chemotherapy: 17.2%
Placebo + chemotherapy: 12.3% Placebo + chemotherapy: 4.3%
Checkmate-77T (14,15) Neoadjuvant nivolumab 360 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant nivolumab 480 mg (q4 weeks for 1 year) vs. neoadjuvant placebo plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant placebo (q4 weeks for 1 year) Resectable stage IIA–IIIB NSCLC; n=461 (n=229 to nivolumab; n=232 to placebo) Nivolumab + chemotherapy: 35.4% Nivolumab + chemotherapy: 25.3%
Placebo + chemotherapy: 12.1% Placebo + chemotherapy: 4.7%
NEOTORCH (16,17) Toripalimab 240 mg plus platinum-based chemotherapy (q3 weeks for 3 cycles before surgery and 1 cycle after surgery) followed by adjuvant toripalimab (q3 weeks for up to 13 cycles) vs. placebo plus platinum-based chemotherapy (3 cycles q3W before surgery and 1 cycle after surgery) followed by adjuvant placebo (q3 weeks for up to 13 cycles) Stage II–III NSCLC; n=404 (n=202 to toripalimab; n=202 to placebo) Toripalimab + chemotherapy: 48.5% Toripalimab + chemotherapy: 24.8%
Placebo + chemotherapy: 8.4% Placebo + chemotherapy: 1.0%

LCMC3, Lung Cancer Mutation Consortium 3 trial; MPR, major pathologic response; PCR, complete pathologic response; NSCLC, non-small cell lung cancer.

Surgical outcomes are summarized in Table 2 (3,7,10-17). Of the 159 patients who underwent surgery, only 4 patients experienced a treatment-related surgical delay. Over half (54%, n=86) underwent a minimally invasive surgery by video-assisted thoracoscopic surgery (VATS) or robotic VATS, and the rate of conversion from minimally invasive surgery to thoracotomy was 15% (n=15) (7). Most patients (79%, n=125) underwent a lobectomy, and pneumonectomy was uncommon (9%, n=14) (7). Complete resection (R0) was achieved in 91% of patients (n=145). Intraoperative complications were rare (n=5) but successfully managed, and the rate of grade 4–5 adverse events (AEs) was higher in patients who received pneumonectomy (28.5%) compared to patients without a pneumonectomy (10%). Pre- and postoperative treatment-related and immune-related AEs (irAEs) were consistent with known immunotherapy-related toxicities.

Table 2

Surgical outcomes in LCMC3 and other neoadjuvant and perioperative trials

Trial Intervention Patient population Rate of surgical resection Rate of minimally invasive surgery Rate of R0 resection
LCMC3 (7) Neoadjuvant atezolizumab 1,200 mg (2 cycles q3 weeks) and adjuvant atezolizumab (up to 12 months) Stage IB–IIIB NSCLC, n=181 (n=143 in efficacy population) 88% 54% 91%
NEOSTAR (10,11) Neoadjuvant nivolumab 3 mg/kg (3 cycles q2 weeks) vs. neoadjuvant nivolumab 3 mg/kg plus ipilimumab 1 mg/kg (3 cycles q2 weeks) Stage I–IIIA NSCLC, n=44 (n=23 to nivolumab; n=21 to nivolumab + ipilimumab) Nivolumab: 91% Nivolumab: 29% Nivolumab: 100%
Nivolumab + ipilimumab: 76% Nivolumab + ipilimumab: 26% Nivolumab + ipilimumab: 100%
KEYNOTE-671 (3) Neoadjuvant pembrolizumab 200 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant pembrolizumab (q3 weeks for up to 13 cycles) vs. neoadjuvant chemotherapy plus placebo (q3 weeks for 4 cycles) and adjuvant placebo (q3 weeks for up to 13 cycles) Stage II–IIIB NSCLC, n=797 (n=397 to pembrolizumab; n=400 to placebo) Pembrolizumab + chemotherapy: 82.1% Not reported Pembrolizumab + chemotherapy: 92%
Placebo + chemotherapy: 79.4% Placebo + chemotherapy: 84.2%
Checkmate-816 (12) Neoadjuvant nivolumab 360 mg plus platinum-based chemotherapy (q3 weeks for 3 cycles) vs. neoadjuvant platinum-based chemotherapy (q3 weeks for 3 cycles) Stage IB–IIIA NSCLC; n=358 (n=179 to nivolumab; n=179 to chemotherapy only) Nivolumab + chemotherapy: 83% Nivolumab + chemotherapy: 29.5% Nivolumab + chemotherapy: 83%
Chemotherapy only: 75.4% Chemotherapy only: 21.5% Chemotherapy only: 78%
AEGEAN (13) Neoadjuvant durvalumab 1,500 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant durvalumab (q4 weeks for up to 12 cycles) vs. neoadjuvant platinum-based chemotherapy plus placebo (q3 weeks for 4 cycles) and adjuvant placebo (q4 weeks for up to 12 cycles) Stage IIA to IIIB NSCLC; n=802 (n=400 to durvalumab; n=402 to placebo) Durvalumab + chemotherapy: 77.6% Durvalumab + chemotherapy: 39.6% Durvalumab + chemotherapy: 94.7%
Placebo + chemotherapy: 76.7% Placebo + chemotherapy: 38.0% Placebo + chemotherapy: 91.3%
Checkmate-77T (14,15) Neoadjuvant nivolumab 360 mg plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant nivolumab 480 mg (q4 weeks for 1 year) vs. neoadjuvant placebo plus platinum-based chemotherapy (q3 weeks for 4 cycles) and adjuvant placebo (q4 weeks for 1 year) Resectable stage IIA–IIIB NSCLC; n=461 (n=229 to nivolumab; n=232 to placebo) Nivolumab + chemotherapy: 78% Not reported Nivolumab + chemotherapy: 89%
Placebo + chemotherapy: 77% Placebo + chemotherapy: 90%
NEOTORCH (16,17) Toripalimab 240 mg plus platinum-based chemotherapy (q3 weeks for 3 cycles before surgery and 1 cycle after surgery) followed by adjuvant toripalimab (q3 weeks for up to 13 cycles) vs. placebo plus platinum-based chemotherapy (3 cycles q3W before surgery and 1 cycle after surgery) followed by adjuvant placebo (q3 weeks for up to 13 cycles) Stage II–III NSCLC; n=404 (n=202 to toripalimab; n=202 to placebo) Toripalimab + chemotherapy: 82.2% Not reported Toripalimab + chemotherapy: 95.8%
Placebo + chemotherapy: 73.3% Placebo + chemotherapy: 92.6%

LCMC3, Lung Cancer Mutation Consortium 3 trial; NSCLC, non-small cell lung cancer.

The encouraging results of LCMC3, the largest single-arm phase II trial of neoadjuvant immunotherapy, provided rationale to conduct subsequent randomized chemoimmunotherapy trials. In addition to size, strengths of the trial include its multicenter design and review of institutional MPR assessments by a central pathology committee (2,7). However, the study has several limitations. First, it is a single-arm study, which limits our ability to draw definitive conclusions regarding the immediate and long-term benefit of neoadjuvant atezolizumab. Second, documentation of lymph node involvement by endobronchial ultrasound or mediastinoscopy for patients with clinical II–IIIA disease was not required, which may lead to inaccurate staging and interfere with the interpretability of results. Third, although surgical candidacy was determined jointly by the treating thoracic surgeon and medical oncologist, criteria for surgical candidacy were not standardized or assessed centrally, which increases the likelihood of selection bias. Fourth, pathologic response was only assessed on primary tumors. Excluding lymph node responses limits the pathologic evaluation and our ability to assess treatment effectiveness (2). Fifth, while MPR is now commonly used as an endpoint for neoadjuvant trials and has been shown to correlate with both overall survival and disease-free survival (19,20), assessing for MPR can be subjective, despite efforts to standardize pathologic calculations (21-23). Finally, most of the patients on the study were white (81%), current or former smokers (90%), and had stage IIB or higher disease (81%), thus limiting the generalizability of results.


Context and implications of pathologic outcomes in LCMC3

The favorable pathologic outcomes observed in LCMC3 provided the basis for administering neoadjuvant immunotherapy to patients with resectable NSCLC. Similar findings were reported in the smaller phase II NEOSTAR trial, which randomized patients to receive neoadjuvant nivolumab or nivolumab plus ipilimumab (Table 1) (10,11). In the wake of LCMC3 and NEOSTAR, several phase III neoadjuvant and perioperative chemoimmunotherapy trials have been conducted and have demonstrated consistently higher levels of pathologic response with the addition of immunotherapy. The control arm across these trials is neoadjuvant chemotherapy alone, which has an established benefit in locally advanced disease but was variably adopted (24-26).

Five large randomized phase III chemoimmunotherapy trials—KEYNOTE-671, Checkmate-816, AEGEAN, Checkmate-77T, and Neotorch—all recently reached their primary endpoints (3,12-14,16). A comparison of pathologic outcomes is summarized in Table 1. Compared to patients treated with neoadjuvant chemoimmunotherapy in the phase III trials, a lower rate of both MPR and pCR was observed in patients in LCMC3. On the other hand, the pathologic response rates observed in LCMC3 appeared to be superior to pathologic response rates in patients treated with chemotherapy only in the phase III trials and to historical rates of pCR after induction chemotherapy (27). There have been no large head-to-head comparisons of immunotherapy alone to either chemoimmunotherapy or chemotherapy alone in the neoadjuvant setting. However, as demonstrated by LCMC3, there may be a subset of patients who benefit from neoadjuvant immunotherapy alone and would not require escalation to chemoimmunotherapy. Although the rate of MPR in LCMC3 was higher in patients with high PD-L1 TPS, no informative biomarkers have been established in the neoadjuvant setting (2). Further investigation is needed to better characterize these subpopulations of patients.


Context and implications of surgical outcomes in LCMC3

Surgical outcomes are a crucial marker of clinical benefit in patients who are treated with curative intent and can have significant impact on quality of life. A comparison of surgical outcomes across trials is summarized in Table 2. At 88%, the rate of surgical resection in LCMC3 was similar to the surgical rate seen with neoadjuvant nivolumab in NEOSTAR and higher than surgical rates observed in the chemoimmunotherapy arms of subsequent phase III trials (11,28). Although there is no clear explanation, this might be due in part to fewer cycles of neoadjuvant treatment and/or the absence of chemotherapy-related AEs in LCMC3. In LCMC3, AEs with neoadjuvant therapy precluded 0.5% of patients (n=1) from proceeding with surgery. The rate of cancelled surgery due to AEs from neoadjuvant chemoimmunotherapy varies between trials from 1.1% in Checkmate-816 and 1.8% in AEGEAN to 3.0% in NEOTORCH and 6.3% in KEYNOTE-671 (3,13,16,28). Although AEs from neoadjuvant therapy appears to be more problematic with chemoimmunotherapy, it is difficult to draw conclusions based on cross-trial comparisons. However, immunotherapy alone inherently carries less toxicity risk than chemoimmunotherapy. For this reason, it may be a reasonable option for patients who are unable to tolerate chemotherapy prior to surgery. Still, further investigation is needed to improve patient selection in this context.

The rate of complete (R0) resection, an important predictor of long-term outcomes, was over 90% in LCMC3 and comparable to chemoimmunotherapy-treated arms of the subsequent phase III trials (Table 2). The rate of minimally invasive surgery, which has implications for patient function and quality of life, was higher in LCMC3 compared to Checkmate-816 and AEGEAN. The high rate of a minimally invasive approach in LCMC3 supports surgical feasibility after immunotherapy, but the discrepancy with the phase III chemoimmunotherapy trials is difficult to interpret. The differences in rate of minimally invasive surgery may be attributed to the increased variability in surgical technique and experience that is seen when conducting larger trials and/or to a larger proportion of patients with stage III disease in Checkmate-816 and AEGEAN. Fifty one percent of patients in LCMC3 had stage IIIA or IIIB disease, compared to 63% of patients in Checkmate-816 with stage IIIA disease, and 71% of patients in AEGEAN with stage IIIA–IIIB disease (7,12,13).

An important consideration when evaluating surgical results is that surgical planning often depends on radiographic assessments, which may not accurately reflect tumor response to immunotherapy. As the authors of LCMC3 note, there was a lack of association between radiologic response and final pathological stage; of the 29 patients with MPR, 25 had stable disease and only 4 had a partial response by RECIST criteria (7). Inaccurate interpretation of tumor response based on imaging studies may affect surgical approach and outcomes across clinical trials that involve neoadjuvant immunotherapy. This highlights the need for better radiographic assessment tools in this setting.

Where we are now? Barriers to adopting neoadjuvant immunotherapy and chemoimmunotherapy

Considering the success of LCMC3 and the recent FDA approvals of neoadjuvant and perioperative chemoimmunotherapy, neoadjuvant and perioperative therapy have become widely, though not universally, adopted. Even though KEYNOTE-671 recently demonstrated a significant improvement in overall survival, some remain hesitant to implement neoadjuvant or perioperative therapy due to a lack of long-term survival benefit thus far (29). Overall survival data from the other randomized studies will be valuable.

While the focus of this commentary is on the neoadjuvant and perioperative approach, adjuvant systemic therapy remains a valid approach especially for patients who proceed to surgery immediately after diagnosis or who are not diagnosed with primary lung malignancy until after surgery. In our opinion, however, neoadjuvant and perioperative approaches are preferred over adjuvant-only treatment for a number of reasons, including the ability to target an intact tumor, assess pathologic response to treatment, and potentially improve surgical resectability (1,30). Adherence to systemic therapy is also likely superior with a neoadjuvant approach, as was previously demonstrated for chemotherapy in the NATCH trial (31). There are no randomized data directly comparing neoadjuvant immunotherapy to adjuvant immunotherapy in patients with lung cancer, but clinical trials in melanoma have demonstrated that moving immunotherapy treatments into the neoadjuvant setting improves outcomes compared to an adjuvant-only approach (32).

Although it is difficult to draw definitive conclusions from cross-trial comparisons, the magnitude of survival benefit observed in neoadjuvant and perioperative trials like KEYNOTE-671 and CheckMate 816 has been consistently greater than what was seen in adjuvant trials such as IMpower010 and KEYNOTE-091 (33,34). Ultimately, we believe the broad spectrum of clinical presentations within resectable NSCLC warrants an equally broad range of treatment options. Investigative efforts aimed at improving patient selection, such as the use of pathologic response to tailor adjuvant treatment, will promote truly personalized medicine in this space.

Surgical risk after neoadjuvant immunotherapy

Additional barriers to implementing neoadjuvant therapy include concerns regarding the consequences of delaying curative resection and the possibility that exposure to systemic therapy increases the risk of surgical complications. The high rates of surgical resection, complete resection, and pathologic response across neoadjuvant trials provide reassurance that delaying surgery for neoadjuvant therapy does not negatively impact clinical outcomes. Furthermore, no new or unexpected surgery-related AEs were reported in either LCMC3 or subsequent phase III chemoimmunotherapy trials. The concern that neoadjuvant immunotherapy increases surgical difficulty and complications has so far not been corroborated in large clinical trials. However, a more detailed report of specific surgical complications may help address these concerns.

Some small early studies of surgical morbidity following neoadjuvant immunotherapy cautioned that dense mediastinal and hilar fibrosis may result after treatment with immunotherapy and lead to more challenging surgical resections (35-37). In a phase I study where 20 patients underwent resection after neoadjuvant nivolumab, 54% of VATS and robotic cases required conversion to thoracotomy, often as a result of hilar inflammation and fibrosis (36). Although concerning, this finding has not been reproduced in larger trials. In NEOSTAR, 40% of operations performed after neoadjuvant nivolumab or nivolumab and ipilimumab were graded by surgeons to be “more complex” than a typical resection, but only one patient required conversion to thoracotomy due to hilar fibrosis (11). The overall perioperative morbidity and mortality in NEOSTAR was thought to be comparable to what has previously been seen with neoadjuvant chemotherapy or upfront resection (11).

Although perihilar fibrosis was not assessed directly in LCMC3, the authors note that the low frequency of intraoperative AEs suggests that fibrosis was not a significant problem (7). Fifteen patients (15%) required conversion to thoracotomy for unclear reasons that may or may not have been related to fibrosis. The rate of conversion to thoracotomy in Checkmate-816 was 11.4% in the nivolumab plus chemotherapy arm and 15.6% in the chemotherapy treated arm, though the exact circumstances are not reported (12). Overall, the current data regarding surgical morbidity and mortality from LCMC3 and phase III chemoimmunotherapy trials are reassuring and do not point towards perihilar fibrosis or other treatment-related surgical complications to be a major concern.

The unique toxicity profile of immune checkpoint inhibitors is important to bear in mind, as surgeons must be aware of potential complications in the perioperative setting. In LCMC3, the overall rate of irAEs was low, and irAEs did not significantly impact surgery (7). Only one patient experienced low-grade pneumonitis prior to surgery, and pulmonary function tests (PFTs), which were performed for all patients before and after neoadjuvant therapy, showed no changes in lung function after immunotherapy (7). In the post-operative setting, 5% of patients (n=8) had pneumonitis (including 3 patients with a grade ≥3 event and one patient who died 2.5 months post-operatively), 3% (n=5) had grade ≥3 colitis, and 4% (n=7) had hypothyroidism.

Long-term toxicities in LCMC3 are not reported, but it is well-known that some irAEs, including many endocrinopathies, can be permanent (38). Patient-reported outcomes (PROs) were not included in LCMC3, though PROs reported from Checkmate-816 and KEYNOTE-671 suggested that adding immunotherapy to chemotherapy does not adversely affect quality of life (39,40). Nevertheless, physicians and patients must acknowledge both the immediate and long-term risk of immunotherapy-related toxicities.


Conclusion

The pathologic and surgical outcomes observed in LCMC3 confirm the benefits of neoadjuvant immunotherapy and mitigate concerns over surgical delay or increased surgical complications. While LCMC3 provides rationale for neoadjuvant immunotherapy and set the stage for subsequent chemoimmunotherapy trials, in this rapidly changing field, studies have demonstrated that neoadjuvant chemoimmunotherapy is the superior approach. Chemoimmunotherapy regimens have achieved the highest pathologic response rates and should be considered over both immunotherapy alone and chemotherapy alone for most patients with locally advanced resectable NSCLC, particularly for high-risk patients (such as those with high tumor burden or with PD-L1 negative tumors) (5).

That being said, LCMC3 remains relevant to clinical practice and to ongoing discussion surrounding patient selection. Twenty percent of patients achieved MPR and 6% of patients pCR with just 2 cycles of neoadjuvant atezolizumab, suggesting that immunotherapy alone likely remains a viable alternative for a select few patients. These patients may derive adequate benefit from immunotherapy alone without being subjected to the added toxicities of chemotherapy. Ongoing efforts should focus on recognizing when de-escalation of care in the neoadjuvant setting is appropriate. This need is even more compelling when considering that chemotherapy-related AEs can preclude a subset of patients from undergoing curative resection. Informative biomarkers are needed to identify those who are vulnerable to chemoimmunotherapy-related AEs and who might still achieve pathologic response with immunotherapy alone.

Finally, although no significant perioperative or intraoperative safety concerns have been reported in LCMC3 or subsequent phase III chemoimmunotherapy trials, a more granular review of intraoperative findings and surgical complications after immunotherapy in these trials is needed. Standardized assessments of perihilar fibrosis and surgical difficulty, for instance, would improve our understanding of the impact of neoadjuvant therapy on curative resection and be informative for patients and multidisciplinary cares teams in the decision-making process.


Future directions

As perioperative management of resectable NSCLC continues to evolve at a rapid pace, many questions remain unanswered but are subject to ongoing investigation. As noted above, informative biomarkers are needed to improve patient selection and decide, for instance, when neoadjuvant immunotherapy alone may be used in place of chemoimmunotherapy. One small previous phase II study, which included 52 patients, found that MPR was higher with chemoimmunotherapy compared to immunotherapy alone among patients with ≥50% PD-L1 expression (41). SKYSCRAPER-05, an ongoing study of neoadjuvant tiragolumab in combination with atezolizumab, with or without chemotherapy in patients with resectable stage II-IIIB disease, includes both a PD-L1 high (≥50%) and all-comer PD-L1 cohort (42). Results of this trial, among other ongoing trials (such as NCT04040361), may provide additional insight on the role of PD-L1 as a biomarker in the neoadjuvant setting.

Whether a neoadjuvant or perioperative approach is superior remains the subject of ongoing debate (43). Although a recent meta-analysis suggests against clinical benefit of adding adjuvant immunotherapy to neoadjuvant chemoimmunotherapy, randomized data are needed (43). It is not clear whether adding adjuvant therapy should depend on pathologic response to neoadjuvant therapy. Does pCR permit the omission of adjuvant therapy, or should the same treatment be continued after surgery given that the disease appears responsive? Should patients without a pathologic response be recommended to undergo additional treatment or further intensified therapy in the adjuvant setting? Circulating tumor DNA to assess minimal residual disease (MRD) may be the key to unlocking the answers. There are several ongoing trials assessing its role in guiding treatment for early-stage NSCLC after definitive therapy (NCT04585477; NCT04585490; NCT04966663) (44,45).

The potential for added benefit to chemoimmunotherapy with additional agents is also under scrutiny. Ongoing trials such as NeoCOAST-2 (NCT05061550), which is investigating the addition of novel agents to a chemoimmunotherapy backbone, and SKYSCRAPER-05 as mentioned above, will continue to push the field forward (46). Targeted therapies are also being increasingly tested in the neoadjuvant setting, as evidenced by studies like NeoADAURA (NCT04351555) and NAUTIKA1 (NCT04302025).

From a global perspective, as neoadjuvant treatment paradigms continue to be revised, regional differences in clinical practice and access to care may become increasingly apparent. A recent report, for instance, discusses regulatory differences between the FDA and European Medicines Agency (EMA) and highlights disparities in patient access and patient care across European countries in the context of novel treatments for resectable early-stage NSCLC (47,48). In addition to differences in regulatory practices, differences in insurance reimbursement structure, other cost considerations, patient population, and clinician preferences may lead to geographic variation in treatment practice. The National Institute for Health and Care Excellence, for instance, recently recommended neoadjuvant nivolumab plus chemotherapy as a cost-effective treatment approach for patients in the United Kingdom (49), Furthermore, since surgical resections are complicated procedures that require trained thoracic surgeons, differences in access to experienced surgical teams also have an impact on outcomes of perioperative therapies. This should all be taken in to consideration when evaluating the global impact of novel therapies in this heterogeneous patient population (48).

Finally, as new treatment approaches emerge for resectable NSCLC, additional information will be needed to guide treatment selection. Real-world studies on treatment effectiveness and incorporation of patient-reported outcomes to ongoing and future clinical trials may provide useful guidance for both patients and practitioners when facing treatment decisions.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, AME Clinical Trials Review. The article has undergone external peer review.

Peer Review File: Available at https://actr.amegroups.com/article/view/10.21037/actr-24-13/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-13/coif). B.P.S. participated in educational talks for Merck. J.R.S. reports consulting fees from Medtronic and served as an advisory board of Genentech. C.A.S. reports personal fees from Arcus Biosciences, AstraZeneca, Daiichi-Sankyo, Genentech, Janssen, Takeda. The other author has no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Shu CA, Cascone T. What Is Neo? Chemoimmunotherapy in the Neoadjuvant Setting for Resectable Non-Small-Cell Lung Cancer. J Clin Oncol 2021;39:2855-8. [Crossref] [PubMed]
  2. Chaft JE, Oezkan F, Kris MG, et al. Neoadjuvant atezolizumab for resectable non-small cell lung cancer: an open-label, single-arm phase II trial. Nat Med 2022;28:2155-61. [Crossref] [PubMed]
  3. Wakelee H, Liberman M, Kato T, et al. Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:491-503. [Crossref] [PubMed]
  4. FDA approves neoadjuvant/ adjuvant pembrolizumab for resectable non-small cell lung cancer [Internet]. 2023. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-neoadjuvant-adjuvant-pembrolizumab-resectable-non-small-cell-lung-cancer
  5. Sorin M, Prosty C, Ghaleb L, et al. Neoadjuvant Chemoimmunotherapy for NSCLC: A Systematic Review and Meta-Analysis. JAMA Oncol 2024;10:621-33. [Crossref] [PubMed]
  6. Jiang J, Wang Y, Gao Y, et al. Neoadjuvant immunotherapy or chemoimmunotherapy in non-small cell lung cancer: a systematic review and meta-analysis. Transl Lung Cancer Res 2022;11:277-94. [Crossref] [PubMed]
  7. Rusch VW, Nicholas A, Patterson GA, 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 ca. J Thorac Cardiovasc Surg 2023;165:828-839.e5. [Crossref] [PubMed]
  8. Lee CK, Man J, Lord S, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non–Small Cell Lung Cancer—A Meta-Analysis. J Thorac Oncol 2017;12:403-7. [Crossref] [PubMed]
  9. Jahanzeb M, Lin HM, Pan X, et al. Immunotherapy Treatment Patterns and Outcomes Among ALK-Positive Patients With Non-Small-Cell Lung Cancer. Clin Lung Cancer 2021;22:49-57. [Crossref] [PubMed]
  10. Cascone T, William WN Jr, Weissferdt A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med 2021;27:504-14. [Crossref] [PubMed]
  11. Sepesi B, Zhou N, William WN Jr, et al. Surgical outcomes after neoadjuvant nivolumab or nivolumab with ipilimumab in patients with non-small cell lung cancer. J Thorac Cardiovasc Surg 2022;164:1327-37. [Crossref] [PubMed]
  12. Forde PM, Spicer J, Lu S, et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med 2022;386:1973-85. [Crossref] [PubMed]
  13. Heymach JV, Harpole D, Mitsudomi T, et al. Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:1672-84. [Crossref] [PubMed]
  14. Cascone T, Awad MM, Spicer JD, et al. LBA1 CheckMate 77T: Phase III study comparing neoadjuvant nivolumab (NIVO) plus chemotherapy (chemo) vs neoadjuvant placebo plus chemo followed by surgery and adjuvant NIVO or placebo for previously untreated, resectable stage II-IIIb NSCLC. Ann Oncol 2023;34:S1295. [Crossref]
  15. Cascone T, Provencio M, Sepesi B, et al. Checkmate 77T: A phase III trial of neoadjuvant nivolumab (NIVO) plus chemotherapy (chemo) followed by adjuvant nivo in resectable early-stage NSCLC. J Clin Oncol 2020;38:TPS9076. [Crossref]
  16. Lu S, Zhang W, Wu 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. [Crossref] [PubMed]
  17. Lu S, Wu L, Zhang W, et al. Perioperative toripalimab + platinum-doublet chemotherapy vs chemotherapy in resectable stage II/III non-small cell lung cancer (NSCLC): Interim event-free survival (EFS) analysis of the phase III Neotorch study. J Clin Oncol 2023;41:425126. [Crossref]
  18. Dacic S, Travis W, Redman M, et al. International Association for the Study of Lung Cancer Study of Reproducibility in Assessment of Pathologic Response in Resected Lung Cancers After Neoadjuvant Therapy. J Thorac Oncol 2023;18:1290-302. [Crossref] [PubMed]
  19. Pataer A, Kalhor N, Correa AM, et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol 2012;7:825-32. [Crossref] [PubMed]
  20. Pataer A, Weissferdt A, Vaporciyan AA, et al. Evaluation of Pathologic Response in Lymph Nodes of Patients With Lung Cancer Receiving Neoadjuvant Chemotherapy. J Thorac Oncol 2021;16:1289-97. [Crossref] [PubMed]
  21. Chen LN, Wei AZ, Shu CA. Neoadjuvant immunotherapy in resectable non-small-cell lung cancer. Ther Adv Med Oncol 2023;15:17588359231163798. [Crossref] [PubMed]
  22. Saqi A, Leslie KO, Moreira AL, 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. [Crossref] [PubMed]
  23. Travis WD, Dacic S, Wistuba I, et al. IASLC Multidisciplinary Recommendations for Pathologic Assessment of Lung Cancer Resection Specimens After Neoadjuvant Therapy. J Thorac Oncol 2020;15:709-40. [Crossref] [PubMed]
  24. Roth JA, Fossella F, Komaki R, et al. A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. J Natl Cancer Inst 1994;86:673-80. [Crossref] [PubMed]
  25. Rosell R, Gómez-Codina J, Camps C, et al. A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small-cell lung cancer. N Engl J Med 1994;330:153-8. [Crossref] [PubMed]
  26. Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 2014;383:1561-71. [Crossref] [PubMed]
  27. Provencio M, Calvo V, Romero A, et al. Treatment Sequencing in Resectable Lung Cancer: The Good and the Bad of Adjuvant Versus Neoadjuvant Therapy. Am Soc Clin Oncol Educ Book 2022;42:1-18. [Crossref] [PubMed]
  28. Forde PM, Chaft JE, Smith KN, et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 2018;378:1976-86. [Crossref] [PubMed]
  29. Spicer JD, Gao S, Liberman M, et al. LBA56 Overall survival in the KEYNOTE-671 study of perioperative pembrolizumab for early-stage non-small-cell lung cancer (NSCLC). Ann Oncol 2023;34:S1297-8. [Crossref]
  30. Topalian SL, Forde PM, Emens LA, et al. Neoadjuvant immune checkpoint blockade: A window of opportunity to advance cancer immunotherapy. Cancer Cell 2023;41:1551-66. [Crossref] [PubMed]
  31. Felip E, Rosell R, Maestre JA, et al. Preoperative chemotherapy plus surgery versus surgery plus adjuvant chemotherapy versus surgery alone in early-stage non-small-cell lung cancer. J Clin Oncol 2010;28:3138-45. [Crossref] [PubMed]
  32. Patel SP, Othus M, Chen Y, et al. Neoadjuvant-Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N Engl J Med 2023;388:813-23. [Crossref] [PubMed]
  33. O'Brien M, Paz-Ares L, Marreaud S, et al. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol 2022;23:1274-86. [Crossref] [PubMed]
  34. Felip E, Altorki N, Zhou C, 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. Lancet 2021;398:1344-57. [Crossref] [PubMed]
  35. Chaft JE, Hellmann MD, Velez MJ, et al. Initial Experience With Lung Cancer Resection After Treatment With T-Cell Checkpoint Inhibitors. Ann Thorac Surg 2017;104:e217-8. [Crossref] [PubMed]
  36. Bott MJ, Yang SC, Park BJ, et al. Initial results of pulmonary resection after neoadjuvant nivolumab in patients with resectable non-small cell lung cancer. J Thorac Cardiovasc Surg 2019;158:269-76. [Crossref] [PubMed]
  37. Bott MJ, Cools-Lartigue J, Tan KS, et al. Safety and Feasibility of Lung Resection After Immunotherapy for Metastatic or Unresectable Tumors. Ann Thorac Surg 2018;106:178-83. [Crossref] [PubMed]
  38. Husebye ES, Castinetti F, Criseno S, et al. Endocrine-related adverse conditions in patients receiving immune checkpoint inhibition: an ESE clinical practice guideline. Eur J Endocrinol 2022;187:G1-G21. [Crossref] [PubMed]
  39. Felip E, Wang C, Ciuleanu T-E, et al. 932MO Nivolumab (NIVO) plus platinum-doublet chemotherapy (chemo) versus chemo as neoadjuvant treatment for resectable non-small cell lung cancer (NSCLC): Health-related quality of life (HRQoL) outcomes from CheckMate 816. Ann Oncol 2022;33:S973-4. [Crossref]
  40. Garassino MC, Wakelee HA, Spicer J, et al. Health-related quality of life (HRQoL) outcomes from the randomized, double-blind phase 3 KEYNOTE-671 study of perioperative pembrolizumab for early-stage non-small-cell lung cancer (NSCLC). J Clin Oncol 2024;42:8012.
  41. Liu SY, Dong S, Yang XN, et al. Neoadjuvant nivolumab with or without platinum-doublet chemotherapy based on PD-L1 expression in resectable NSCLC (CTONG1804): a multicenter open-label phase II study. Signal Transduct Target Ther 2023;8:442. [Crossref] [PubMed]
  42. Pass H, Kim AW, Felip E, et al. 970TiP SKYSCRAPER-05: Phase II study of neoadjuvant atezolizumab (Atezo) + tiragolumab (Tira) with or without platinum-based chemotherapy (CT) in patients (Pts) with locally advanced resectable stage II-IIIB NSCLC. Ann Oncol 2022;33:S990-1. [Crossref]
  43. Zhou Y, Li A, Yu H, et al. Neoadjuvant-Adjuvant vs Neoadjuvant-Only PD-1 and PD-L1 Inhibitors for Patients With Resectable NSCLC: An Indirect Meta-Analysis. JAMA Netw Open 2024;7:e241285. [Crossref] [PubMed]
  44. 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;28:3308-17. [Crossref] [PubMed]
  45. Bestvina CM, Garassino MC, Neal JW, et al. Early-Stage Lung Cancer: Using Circulating Tumor DNA to Get Personal. J Clin Oncol 2023;41:4093-6. [Crossref] [PubMed]
  46. Guisier F, Bennouna J, Spira AI, et al. NeoCOAST-2: A phase 2 study of neoadjuvant durvalumab plus novel immunotherapies (IO) and chemotherapy (CT) or MEDI5752 (volrustomig) plus CT, followed by surgery and adjuvant durvalumab plus novel IO or volrustomig alone in patients with resectable non-s. J Clin Oncol 2023;41:TPS8604. [Crossref]
  47. Houda I, Dickhoff C, Uyl-de Groot CA, et al. New systemic treatment paradigms in resectable non-small cell lung cancer and variations in patient access across Europe. Lancet Reg Health Eur 2024;38:100840. [Crossref] [PubMed]
  48. Houda I, Dickhoff C, Uyl-de Groot CA, et al. Challenges and controversies in resectable non-small cell lung cancer: a clinician's perspective. Lancet Reg Health Eur 2024;38:100841. [Crossref] [PubMed]
  49. Nivolumab with chemotherapy for neoadjuvant treatment of resectable non-small-cell lung cancer. National Institute for Health and Care Excellence. 2023. Available online: www.nice.org.uk/guidance/ta876
doi: 10.21037/actr-24-13
Cite this article as: Chen LN, Stanifer BP, Sonett JR, Shu CA. Advancing neoadjuvant immunotherapy in non-small cell lung cancer (NSCLC): commentary on the Lung Cancer Mutation Consortium 3 trial (LCMC3). AME Clin Trials Rev 2024;2:37.

Download Citation