2022 WCLC Highlights

CME

Independent Conference Coverage of the IASLC 2022 World Conference on Lung Cancer

Physicians: Maximum of 1.00 AMA PRA Category 1 Credit

Released: October 25, 2022

Expiration: October 24, 2023

Wade T. Iams
Wade T. Iams, MD, MSCI
Heather Wakelee
Heather Wakelee, MD

Activity

Progress
1
Course Completed
CALGB 140503 (Alliance): Background

Heather Wakelee, MD:
One of the studies that was highlighted at WCLC 2022 was the phase III CALGB 140503 (Alliance) trial, which was highly anticipated in the surgical world to help determine whether less extensive surgery in patients with early-stage NSCLC can be considered.1 Instead of the standard full lobar resection, the investigators examined whether sublobar resection would be equivalent for patients who had small tumors. The hope was that we would be able to preserve more lung function with this approach.

Recently, the phase III JCOG0802/WJOG4607L trial from Japan showed significant OS benefit by doing segmentectomy vs lobectomy in patients who had small peripheral stage IA NSCLC with a tumor diameter ≤2 cm.2

CALGB 140503 (Alliance): Study Design

 
Heather Wakelee, MD:
The phase III CALGB 140503 (Alliance) study enrolled patients with peripheral NSCLC T1aN0 with a tumor size ≤2 cm, with pathologically confirmed lymph node negative status at level 10 and mediastinum (2 stations). The patients were randomized to receive lobectomy or sublobar resection, which could be either segmentectomy or wedge resection. The primary endpoint was disease-free survival (DFS).1

 

CALGB 140503 (Alliance): Baseline Characteristics

 
Heather Wakelee, MD:
The trial randomized almost 700 patients in total, with fairly well-balanced baseline characteristics across the arms. Most patients had a smoking history or were active current smokers, and very few patients had never smoked. Also, a majority of the patients had nonsquamous histology.1

CALGB 140503 (Alliance): DFS (Primary Endpoint)

 
Heather Wakelee, MD:
The primary endpoint of DFS was met in this noninferiority trial. For patients with small tumors and node-negative NSCLC, whether they had a full lobectomy or sublobectomy with segmentectomy or wedge resection did not impact the DFS. The HR for DFS was 1.01 (90% CI: 0.83-1.24), with a 1-sided P = .0176 from a noninferiority test. The 5-year DFS was similar at 64.1% with a lobectomy vs 63.6% with a sublobar resection.1

CALGB 140503 (Alliance): DFS by Subgroup

 
Heather Wakelee, MD:
The investigators also assessed DFS by various subgroups and these generally showed the same trend in DFS as the overall patient population. There were a few outliers, such as patients with a right middle lobe tumor location or patients who had never smoked, but these were very small subgroups, which makes data analysis difficult. Overall, these data suggest that less extensive surgery with a sublobar resection did seem to be equivalent to a lobectomy in most patient subgroups.1

CALGB 140503 (Alliance): OS and NSCLC-Related Events vs Competing Deaths

 
Heather Wakelee, MD:
Similar to the results with DFS, there was also no difference in OS between patients who received a full lobectomy or a sublobectomy. These results differ from the JCOG0802/WJOG4607L trial, which showed an OS benefit with segmentectomy vs lobectomy.2

In this trial so far, we are not seeing an OS benefit but there was also not a decrement in OS.1 There was also no appreciable difference in lung cancer–related events vs competing deaths when comparing lobar vs sublobar resection in this study.

CALGB 140503 (Alliance): Disease Recurrence and Pulmonary Function

 
Heather Wakelee, MD:
So why do a smaller resection vs a lobectomy if there is no clear difference in survival outcomes? The advantage for sublobar resection is the potential for better preservation of lung function. In this trial, the investigators carefully assessed pulmonary function as part of the trial, including an assessment of forced expiratory volume in 1 second (FEV1) and forced vital capacity (FVC) at baseline and then again at 6 months after surgery.

They were able to show a statistically significant benefit for sublobar resection with a smaller decrease in both FEV1 and FVC from baseline vs with a lobectomy, meaning more pulmonary function was preserved with a sublobar resection.1

CALGB 140503 (Alliance): Conclusions

Heather Wakelee, MD:
The most important aspect of this second trial comparing lobectomy vs sublobar resection, done predominantly in North America, was that it confirmed that the DFS and OS were not inferior with a sublobar resection vs a lobectomy in patients with stage IA NSCLC with a tumor diameter ≤2 cm and without any nodal metastases and that sublobar resection has the potential for better preservation of lung function.

There was a lot of discussion about this trial after WCLC 2022, with many people saying that this trial helped establish a new standard of care, or at least an option to discuss sublobar resection with patients in this particular subgroup. 

Wade T. Iams, MD, MSCI:
I would just note that surgeons at my institution are very excited about these data. Thinking about these data in the context of a tumor board, for a patient with a 2 cm or smaller primary tumor and no nodal involvement who is a borderline surgical candidate, these data show that sublobar resection may be an option for them. In the past, we were not certain whether their survival would be similar to doing a lobectomy, so these data help reinforce that this is a good option for these patients.

NADIM II: Background

Wade T. Iams, MD, MSCI:
For our patients with early-stage NSCLC, not only is the type of resection important, but the risks and benefits for perioperative systemic therapy should also be considered. The randomized phase II NADIM II study assessed neoadjuvant nivolumab plus chemotherapy in patients with locally advanced, potentially resectable stage IIIA/B NSCLC.3 We do have a recent FDA approval of neoadjuvant nivolumab plus chemotherapy for resectable, early-stage NSCLC based on the phase III CheckMate 816 data.4 However, we do not yet have OS data from CheckMate 816, although there was a significantly longer event-free survival and a higher percentage of patients with a pathological CR with nivolumab plus chemotherapy than with chemotherapy alone. This analysis from NADIM II allows us to look at a good dataset with more mature results to start exploring OS results for this treatment option.

NADIM II: Study Design

 
Wade T. Iams, MD, MSCI:
NADIM II enrolled 86 patients with potentially resectable stage IIIA/B NSCLC who had no sensitizing EGFR or ALK alterations. Patients were randomized to receive neoadjuvant nivolumab plus chemotherapy with paclitaxel and carboplatin or chemotherapy alone every 3 weeks for 3 cycles, followed by surgery 3-4 weeks after Day 21 of the third cycle of neoadjuvant therapy, and then adjuvant nivolumab every 2 weeks for 6 months or observation, respectively. The primary endpoint was the pathologic CR (pCR) rate.3

 

NADIM II: Baseline Characteristics (ITT)

 
Wade T. Iams, MD, MSCI:
Baseline characteristics were generally well balanced. The incidence of adenocarcinoma and squamous cell carcinoma was fairly balanced in the patient population, although there were slightly more patients with adenocarcinoma in the nivolumab arm. The majority of patients had N2 disease at inclusion onto this study, but again, there were slightly more patients with N2 disease in the nivolumab arm. Overall, the patients enrolled in this trial represent a higher‑risk patient population.3

NADIM II: Surgery Summary

 
Wade T. Iams, MD, MSCI:
It is always a concern that embarking on systemic neoadjuvant therapy could confound the surgical options for patients, but it looks like that was not the case in this study. In fact, patients were able to reach a definitive surgery significantly more often with nivolumab plus chemotherapy vs chemotherapy alone (93% vs 69% of patients; P = .04), so it was a reassuring finding.3

NADIM II: Downstaging (Secondary Endpoint)

 
Wade T. Iams, MD, MSCI:
Looking at patients who achieved downstaging at surgery, there was a benefit from the addition of neoadjuvant nivolumab to chemotherapy. Almost 70% of patients receiving nivolumab plus chemotherapy achieved downstaging at surgery compared with 40% with chemotherapy alone.3

NADIM II: PFS (Secondary Endpoint)

 
Wade T. Iams, MD, MSCI:
Of importance, the addition of nivolumab to chemotherapy significantly improved the median progression‑free survival (PFS) compared with those who received neoadjuvant chemotherapy alone (not reached vs 18.3 months; HR: 0.48; P = .025).3 

NADIM II: PFS by pCR Status (Secondary Endpoint)

 
Wade T. Iams, MD, MSCI:
There is a lot of discussion about the use of pCR as a surrogate endpoint before we have survival results for therapy approval. These data suggest that patients who achieved a pCR had a greatly improved PFS compared with those who had an incomplete pathological response.3 

NADIM II: OS (Secondary Endpoint)

 
Wade T. Iams, MD, MSCI:
This analysis focused on OS, and showed an impressive OS rate at 1 year among patients who received neoadjuvant nivolumab plus chemotherapy at 98% compared with 82% in patients who received chemotherapy alone. This was continued at 2 years, with an OS rate of 85% vs 63%, respectively.3 The HR for OS was 0.40 (95% CI: 0.17-0.93) with P = .034. These data provide some impressive survival numbers to include in a discussion about adding neoadjuvant nivolumab to chemotherapy with high‑risk patients.

NADIM II: OS by pCR Status (Secondary Endpoint)

 
Wade T. Iams, MD, MSCI:
Similar to PFS, OS was dramatically improved in patients who achieved a pCR vs those who did not, regardless of the therapy received.3 As far as using pCR as a surrogate endpoint, it is good to see that among the smaller cohort of patients who received chemotherapy and achieved a pCR (2 of 28 patients), they also seem to have a dramatically improved OS.

NADIM II: Conclusions

Wade T. Iams, MD, MSCI:
These data from NADIM II solidify the recent FDA approval for neoadjuvant nivolumab added to platinum doublet chemotherapy for eligible patients with resectable NSCLC tumors (≥4 cm or node positive), based on the CheckMate 816 study. The data also provide some additional context to this important practice change with excellent survival results.

Heather Wakelee, MD:
NADIM II is not necessarily practice changing because we already had CheckMate 816, which established the addition of neoadjuvant nivolumab to chemotherapy; however, Dr. Iams highlighted a couple of the key features from this trial. I think it is important to remember that this approach should not be considered for all patients; the trial excluded patients who had driver alterations, specifically EGFR mutations or ALK translocations, in their tumor and only included stage III patients. Some healthcare professionals believe that a patient with stage IIIA and especially IIIB NSCLC should not have surgery but should receive concurrent chemoradiation plus additional immune therapy with durvalumab. However, these impressive OS results with neoadjuvant nivolumab added to chemotherapy along with some of the other impressive outcomes of this trial, including pCR rates, do make us pause and they will make our tumor boards continue to have lots of discussions.

When considering both the CheckMate 816 and NADIM II trials, there were some differences to keep in mind. NADIM II only used carboplatin/paclitaxel as chemotherapy, whereas CheckMate 816 allowed various chemotherapy regimens, including cisplatin/pemetrexed or carboplatin/paclitaxel for patients with nonsquamous histology and cisplatin/gemcitabine or carboplatin/paclitaxel for squamous histology. In addition, patients in the chemotherapy-only arm or patients in either treatment group receiving adjuvant chemotherapy could also receive vinorelbine/cisplatin or docetaxel/cisplatin. NADIM II confirmed that using a taxane-based platinum doublet can be a good strategy and lends support for these neoadjuvant strategies.

NADIM II also continued nivolumab in the adjuvant setting for those in the nivolumab arm, while CheckMate 816 offered optional adjuvant chemotherapy for both arms. Now, one of our bigger unanswered questions is the importance of continuing immunotherapy after resection as adjuvant therapy vs only as a part of neoadjuvant therapy, as was done in CheckMate 816.

IMpower010: Background

Heather Wakelee, MD:
The Presidential Plenary session at WCLC 2022 was focused on early‑stage lung cancer because we had some great data from new trials and updated trials. One of the trial updates was from IMpower010, which was the first randomized phase III trial to prove a benefit with adjuvant immune checkpoint inhibitor therapy.

The results from IMpower010 were initially presented at the 2021 American Society of Clinical Oncology (ASCO) annual meeting and showed a significant DFS improvement with adjuvant atezolizumab vs BSC for patients with completely resected stage IB-IIIA NSCLC after platinum-based chemotherapy.5 Based on these data, atezolizumab was approved as adjuvant treatment following resection and platinum-based chemotherapy for adult patients with stage II-IIIA NSCLC with PD-L1 expression on ≥1% of tumor cells in countries including the United States, China, and Japan.

The updates at WCLC 2022 included a prespecified interim analysis of the OS events. 

IMpower010: Study Design

 
Heather Wakelee, MD:
IMpower010 was a randomized, open-label phase III trial that included patients who had completely resected stage IB‑IIIA NSCLC, with tumors ≥4 cm in size if it was IB. Patients received adjuvant chemotherapy with cisplatin plus pemetrexed, gemcitabine, docetaxel, or vinorelbine. They were then randomized to either atezolizumab every 3 weeks for 16 cycles or BSC. The primary endpoint was hierarchical evaluation of investigator-assessed DFS.6 OS was a secondary endpoint to be assessed if the primary endpoints were positive.

IMpower010: OS in Patients With Stage II-IIIA NSCLC and PD-L1 TC ≥1%

 
Heather Wakelee, MD:
In the FDA approved patient population with stage II‑IIIA NSCLC and PD‑L1 ≥1%, the HR for OS was 0.71 (95% CI: 0.49-1.03) at a median follow-up of 46 months.6 We cannot call that statistically significant at this point; however, at the last data cut, the HR for OS in this patient population was 0.77.5 These 2 analyses suggest that the survival curves are continuing to separate more overtime.

Additional follow-up is needed to determine if a significant difference in OS emerges, but these data suggest that it is trending in that direction. These data were reassuring to many who have been questioning whether there will actually be a survival benefit. Looking at the OS curve separation at 3 years, 82% of patients remain alive with atezolizumab vs 79% with BSC. When you get out toward 5 years, more patients are censored, leading to some instability of the numbers, but 77% of patients remain alive with atezolizumab vs 68% with BSC.6

IMpower010: OS in Other Patient Subgroups

 
Heather Wakelee, MD:
In the patient subgroups of all randomized patients with stage II‑IIIA disease, including the 45% of patients without an appreciable PD‑L1 expression, there was absolutely no separation of the OS curves, with an HR of 0.95. This is also true in the intention-to-treat population with the addition of the 12% of patients with stage IB disease; again, the OS curves were similar with or without atezolizumab, with an HR of 0.995.6

IMpower010: OS in Stage II-IIIA NSCLC With PD-L1 TC ≥1% Across Key Subgroups

 
Heather Wakelee, MD:
When we look at the forest plot of OS in patients with stage II‑IIIA NSCLC and PD-L1 expression ≥1%, most groups show a similar benefit with atezolizumab.6 Subgroups that did not trend in favor of atezolizumab included patients whose tumors have an ALK translocation and, potentially, those with an EGFR mutation. In patients with an EGFR mutation, there seemed to be some benefit, but the CI crossed 1.

These patient subgroups are small, but it highlights the need to use biomarker testing, even in early-stage disease, to make sure that we are not including patients with ALK translocations (and probably ROS1 alterations) or EGFR mutations for treatment with immune checkpoint inhibitors in the perioperative setting.

IMpower010: OS By Biomarker Status (Stage II-IIIA)

 
Heather Wakelee, MD:
In a separate subgroup analysis of patients with stage II-IIIA disease and various PD‑L1 expression levels with or without the group of patients with driver mutations, we see that the group who benefits the most from atezolizumab are the patients with ≥50% of tumors expressing PD‑L1. The HR shifts slightly when including patients with EGFR/ALK driver alterations (HR: 0.43 for patients with ≥50% of tumors expressing PD‑L1 when including EGFR/ALK-positive patients vs 0.42 when excluding EGFR/ALK-positive patients).

For patients with 1% to 49% of tumor cells expressing PD-L1, the HR approaches 1 and it is not clear whether there is any benefit. Furthermore, in patients who do not have PD‑L1 expression on their tumors (<1% of tumor cells), there is actually potential harm with an HR of 1.36 when including EGFR/ALK-positive patients or 1.21 when excluding EGFR/ALK-positive patients.

When we look just at the stage II-IIIA patient population with high PD‑L1 expression excluding the EGFR- and ALK-positive population, the separation of OS curves is quite striking.6 Again, at this point we cannot call this statistically significant based on the way the study was powered, but we do see a trend toward OS improvement.

IMpower010: Safety

 
Heather Wakelee, MD:
There were not any new safety signals associated with adjuvant atezolizumab in this longer follow-up. Overall, safety events occurred at similar rates when comparing the median follow-up of 32 months vs 46 months.6

IMpower010: Conclusions

Heather Wakelee, MD:
The current analysis of IMpower010 includes data at nearly 4 years of follow-up and suggests a trend toward improvement in OS with adjuvant atezolizumab, with the OS curves continuing to separate. Currently, there is not a statistically significant difference in OS at this time, and we need to continue to analyze this trial with longer follow-up. 

Wade T. Iams, MD, MSCI:
These data are important to continue to assess for this FDA-approved regimen of adjuvant atezolizumab in patients with ≥1% PD‑L1 expression. The OS results can help solidify the practice in patients whose tumors have ≥50% PD‑L1 expression and the separation of the OS curves is impressive.

In the subgroup of patients with 1% to 49% PD-L1 expression, the benefit of adjuvant atezolizumab has been questioned and it remains a question based on these data. As the data continue to mature, it will be helpful to see if there continues to be a difference in the groups of patients with ≥50% vs 1% to 49% PD-L1 expression.

There is also another subgroup analysis that I will be interested to watch over time, particularly in patients with 1% to 49% PD-L1 expression where the benefit from adjuvant atezolizumab hasn’t been as clear—using circulating tumor DNA (ctDNA) to define measurable residual disease (MRD) negativity. There was an interesting early analysis from the IMpower010 data based on ctDNA results and, potentially, patients with ≥1% PD-L1 expression who are ctDNA positive may show a more pronounced DFS improvement with adjuvant atezolizumab vs placebo compared with the ctDNA-negative population.7 I look forward to seeing more mature data from IMpower010 as the MRD field matures.

In the phase III IMpower010 trial, the largest trend towards an overall survival (OS) benefit with adjuvant atezolizumab vs best supportive care (BSC) in patients with completely resected stage II-IIIA NSCLC was seen in which of the following subpopulations by PD-L1 expression at the first OS interim analysis?