Neoadjuvant Nivolumab Shows Long-Term Benefit in Lung Cancer Patients


Lung Cancer Illustration

According to a new study in the journal Clinical Cancer Research, patients with non-small cell lung cancer, the most common type of lung cancer, who were treated with neoadjuvant nivolumab had improved five-year recurrence-free and overall survival rates.

Study reports five-year survival outcomes.

Patients with resectable non-small cell lung cancer (NSCLC) who were treated with neoadjuvant nivolumab had improved five-year recurrence-free and overall survival rates compared with historical outcomes.

The research will be published today, February 15, 2023, in Clinical Cancer Research, a journal of the American Association for Cancer Research (AACR), a non-profit organization dedicated to advancing cancer research and improving patient outcomes through education, collaboration, and advocacy..

Patrick Forde, MBBCh, the senior author of the study, is an associate professor of oncology and director of the Thoracic Oncology Clinical Research Program at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins.

Samuel Rosner, MD, is co-first author of the study and is a medical oncology fellow at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and a member of Forde’s research group.

NSCLC Background

NSCLC is the most common type of lung cancer and is a leading cause of cancer-related death worldwide. Despite strides in treating metastatic NSCLC, new treatments for earlier-stage disease have only recently emerged, according to Forde.

Rosner added that there is great interest in optimizing neoadjuvant strategies for earlier-stage NSCLCs that are eligible for surgical resection. Rosner is a medical oncology fellow at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and a member of Forde’s research group.

Forde, Rosner, and colleagues previously reported safety and efficacy results from a phase II clinical trial in which patients with stage I-III resectable NSCLC were treated with two doses of neoadjuvant nivolumab. Major pathological responses were observed in 45 percent of patients, independent of tumor PD-L1 expression, and 73 percent of patients whose tumors were surgically resected were recurrence-free 18 months following surgery.

The latest publication reports the final analyses from this trial, including five-year recurrence-free and overall survival rates for the 20 patients who underwent surgical resection.

“To our knowledge, this is the longest follow-up to date for a PD-1/PD-L1 inhibitor in the neoadjuvant setting for any solid tumor,” said Forde.

Study Results

Among the 20 patients who underwent surgical resection, 12 patients (60 percent) remained recurrence-free five years after surgery, and 16 patients (80 percent) were alive, exceeding the 36 to 68 percent five-year survival rate historically observed for patients with stage I-III NSCLC, Rosner noted. Forde added that the observed patient outcomes after neoadjuvant nivolumab were better than those historically observed among patients treated with neoadjuvant chemotherapy.

The authors also identified major pathologic response after neoadjuvant nivolumab as a potential predictive biomarker of recurrence-free and overall survival. Of the nine patients who had a major pathological response after neoadjuvant nivolumab, eight were alive and cancer-free five years after treatment. One patient experienced a recurrence within the first 10 months after treatment but has since been disease-free after definitive chemoradiation. The one death in this subgroup was unrelated to cancer.

In contrast, six of the 11 patients who did not have a major pathological response experienced disease recurrence, and three of these patients died due to their cancer. These results indicate that a major pathological response following neoadjuvant nivolumab may be associated with a lower risk of disease recurrence and death, although the authors caution that these results are preliminary and require further validation in larger studies.

Neoadjuvant nivolumab did not lead to surgical delays, and there was only one late-onset immune-related adverse event, which occurred 16 months after nivolumab treatment and was successfully managed, the authors noted.

“The results from the five-year follow-up analysis indicate that neoadjuvant nivolumab was safe in long-term follow-up and led to encouraging survival in this patient cohort,” said Forde. “The long-term safety and efficacy data from this study provide further support for the use of nivolumab in the neoadjuvant setting.”

Neoadjuvant nivolumab in combination with chemotherapy was approved by the U.S. Food and Drug Administration in March 2022 for the treatment of lung cancer. “Further studies will help us determine whether select patients may benefit from immunotherapy alone,” Forde noted.

“An interesting finding from the analysis was the difference in outcomes between patients with and without a major pathological response,” said Rosner. “Although the sample size was small, the results illustrate the potential power of pathological response as a predictive biomarker.”

Reference: “Five-Year Clinical Outcomes after Neoadjuvant Nivolumab in Resectable Non-Small Cell Lung Cancer” by Samuel Rosner, Joshua E. Reuss, Marianna Zahurak, Jiajia Zhang, Zhen Zeng, Janis Taube, Valsamo Anagnostou, Kellie N. Smith, Joanne Riemer, Peter B. Illei, Stephen R. Broderick, David R. Jones, Suzanne L. Topalian, Drew M. Pardoll, Julie R. Brahmer, Jamie E. Chaft and Patrick M. Forde, 16 February 2023, Clinical Cancer Research.
DOI: 10.1158/1078-0432.CCR-22-2994

Nivolumab for Patients With High-Risk Oral Leukoplakia:A Nonrandomized Controlled Trial


Key Points

Question  Can immune checkpoint therapy treat high-risk oral precancerous disease to prevent progression to oral squamous carcinoma?

Findings  This phase 2 nonrandomized controlled trial treated 33 patients with high-risk oral proliferative verrucous leukoplakia with the programmed cell death 1 protein inhibitor nivolumab and demonstrated variable lesion regression by size and degree of dysplasia in response to therapy, while 27% of patients developed invasive oral cancer after nivolumab. All whole-exome sequenced patients who progressed to develop cancer had 9p21.3 chromosomal loss.

Meaning  Nivolumab showed potential clinical activity in this immune checkpoint therapy trial for high-risk oral precancerous disease; future trials should prioritize cancer-free survival end points and biomarker stratification.

Abstract

Importance  Proliferative verrucous leukoplakia (PVL) is an aggressive oral precancerous disease characterized by a high risk of transformation to invasive oral squamous cell carcinoma (OSCC), and no therapies have been shown to affect its natural history. A recent study of the PVL immune landscape revealed a cytotoxic T-cell–rich microenvironment, providing strong rationale to investigate immune checkpoint therapy.

Objective  To determine the safety and clinical activity of anti–programmed cell death 1 protein (PD-1) therapy to treat high-risk PVL.

Design, Setting, and Participants  This nonrandomized, open-label, phase 2 clinical trial was conducted from January 2019 to December 2021 at a single academic medical center; median (range) follow-up was 21.1 (5.4-43.6) months. Participants were a population-based sample of patients with PVL (multifocal, contiguous, or a single lesion ≥4 cm with any degree of dysplasia).

Intervention  Patients underwent pretreatment biopsy (1-3 sites) and then received 4 doses of nivolumab (480 mg intravenously) every 28 days, followed by rebiopsy and intraoral photographs at each visit.

Main Outcomes and Measures  The primary end point was the change in composite score (size and degree of dysplasia) from before to after treatment (major response [MR]: >80% decrease in score; partial response: 40%-80% decrease). Secondary analyses included immune-related adverse events, cancer-free survival (CFS), PD-1 ligand 1 (PD-L1) expression, 9p21.3 deletion, and other exploratory immunologic and genomic associations of response.

Results  A total of 33 patients were enrolled (median [range] age, 63 [32-80] years; 18 [55%] were female), including 8 (24%) with previously resected early-stage OSCC. Twelve patients (36%) (95% CI, 20.4%-54.8%) had a response by composite score (3 MRs [9%]), 4 had progressive disease (>10% composite score increase, or cancer). Nine patients (27%) developed OSCC during the trial, with a 2-year CFS of 73% (95% CI, 53%-86%). Two patients (6%) discontinued because of toxic effects; 7 (21%) experienced grade 3 to 4 immune-related adverse events. PD-L1 combined positive scores were not associated with response or CFS. Of 20 whole-exome sequenced patients, all 6 patients who had progression to OSCC after nivolumab treatment exhibited 9p21.3 somatic copy-number loss on pretreatment biopsy, while only 4 of the 14 patients (29%) who did not develop OSCC had 9p21.3 loss.

Conclusions and Relevance  This immune checkpoint therapy precancer nonrandomized clinical trial met its prespecified response end point, suggesting potential clinical activity for nivolumab in high-risk PVL. Findings identified immunogenomic associations to inform future trials in this precancerous disease with unmet medical need that has been difficult.

Introduction

Oral leukoplakia refers to a white plaque of variable cancer risk, having excluded other conditions, and affects up to 5% of the global population,1 but only a small proportion of leukoplakia lesions will undergo malignant transformation.2 Degree of epithelial dysplasia, lesion size, and tobacco history all influence the transformation rate.3 Proliferative verrucous leukoplakia (PVL) defines an aggressive subtype with a malignant transformation rate exceeding 10% per year, characterized by heterogeneous or verrucous lesions involving multiple oral subsites.46 To date, no therapies have been shown to change the natural history of this severe oral precancerous disease,79 reflecting a critical unmet medical need.

Studies of the immune landscape led to pivotal trials of anti–programmed cell death 1 protein (PD-1) therapy in recurrent/metastatic head and neck squamous cell carcinoma.1013 Our prior retrospective study revealed a cytotoxic T-cell–rich immune microenvironment in PVL.14 These findings together with immunosurveillance studies in the context of lung premalignancy and of various immune-oncology interventions in preclinical models1518 provided strong rationale for investigating PD-1/PD-1 ligand 1 (PD-L1) axis blockade in oral precancerous disease. Here we report the first (to our knowledge) trial to evaluate the safety and clinical activity of preventive anti–PD-1 therapy among patients with high-risk PVL.

Methods

Study Population

This was an open-label, single-group phase 2 trial conducted at the Dana-Farber Cancer Institute in Boston, Massachusetts (trial protocol in Supplement 1). Patients with high-risk oral leukoplakia defined by any of the following criteria were eligible: PVL with multifocal (≥2), contiguous 3 cm or greater, or a single lesion 4 cm or greater in largest diameter (2-3-4 rule) with epithelial dysplasia (any degree); PVL with 4-quadrant oral cavity involvement; at least 1 localized leukoplakia with moderate dysplasia, or erythroleukoplakia for which surgery was indicated but not feasible or the patient refused. Patients were 18 years or older and had an Eastern Cooperative Oncology Group performance status of 2 or lower. A history of surgically treated carcinoma in situ (CIS) or early-stage oral squamous cell carcinoma (OSCC) (American Joint Committee on Cancer Staging Manual, eighth edition, stages I or II) was permitted. Participants defined their race and ethnicity by self-identification. This was assessed given the potential for variation in interpreting the results of the study based on a majority of participants from 1 ethnic group and/or race given the epidemiology of the disease and the treatment center’s regional participant demographics. The trial was approved by the Dana-Farber/Harvard Cancer Center institutional review board (18-387), conducted in accordance with the Declaration of Helsinki and Good Clinical Practice Guidelines, and registered nationally (NCT03692325). This study followed the Transparent Reporting of Evaluations With Nonrandomized Designs (TREND) reporting guideline.

Treatment

Following written informed consent, participants received nivolumab (480 mg intravenously) on day 1 of a 28-day cycle for 4 cycles. Immunosuppressive medications and doses of corticosteroids greater than 20-mg prednisone equivalent daily were prohibited unless used for immune-related toxicity management.

Assessments

Three weeks prior to the first dose of nivolumab and at monthly visits, patients underwent digital intraoral color photography to capture all leukoplakia lesions. Bidimensional measurements were obtained from up to 3 target lesions (per patient) as determined by 1 of 5 oral medicine investigators. Screening and posttreatment biopsies were performed by the same oral medicine investigator for consistency. Fresh tissue biopsies from all target lesions were mandatory at baseline and 30 days after the final dose of nivolumab. Pathologic specimens from each biopsy were examined by 2 experienced oral pathologists (V.Y.J. and K.S.W.) blinded to outcome data (or a third in cases of any scoring discrepancy). New or suspicious nontarget lesions or changes in target lesions could trigger rebiopsy at any point.

Response was assessed according to a modified composite scoring system (van der Waal classification)19 (eFigure 1 in Supplement 2). The sum of target lesion point scores (both clinical and pathologic) yielded a composite score. The percent change in composite score before and after treatment determined best overall response. Major response (MR) was a decrease of more than 80%, partial response (PR) a decrease of 40% to 80%, stable disease (SD) was neither an MR or PR, and progression of disease (PD) was defined as an increase of 10% or greater in the composite score or a CIS or OSCC diagnosis. Patients were followed up with clinical examinations every 3 to 4 months until study withdrawal or up to 5 years.

Safety

Safety evaluations included laboratory and adverse event (AE) assessments (National Cancer Institute Common Terminology Criteria for Adverse Events, version 5.0).20 For patients who developed grade 3 or intolerable grade 2 immune-related AEs (irAEs), nivolumab treatment could be interrupted, delayed, or discontinued; certain grade 4 irAEs required discontinuation. AEs were captured up to 3 months after completion of nivolumab treatment.

Statistical Analysis

The primary end point was best overall response (MR + PR rate) as defined by the percent change in clinical-pathologic composite score. A 2-stage Simon optimal design was used. When more than 5 of 33 patients who were eligible and began protocol treatment had disease in response (assuming >1 patient with disease in response among the first 16 patients), there was 84.3% power to rule out a 10% and detect a 25% response rate (using a 1-sided exact binomial test, type I error rate of 10%). A response rate of 25% was targeted when considering the cumulative risk of serious irAEs.21

Secondary end points included safety and cancer-free survival (CFS), defined as the time from trial registration to OSCC or death due to any cause (participants alive without oral cancer were censored at last assessment). Based on studies of PD-L1 expression14,22 and somatic 9p21 copy-number loss in advanced OSCC, lung, and other tumors,23,24 we conducted secondary analyses to evaluate the association of pretreatment dysplastic tissue PD-L1 expression and 9p21.3 deletion status with outcomes. Exploratory analyses included immunogenomic profiling using multiparametric flow cytometry and whole-exome sequencing (WES) detailed in the eMethods in Supplement 2.

The primary clinical activity population included all eligible patients who began protocol treatment. Response rate was summarized as a proportion with a corresponding 2-stage 95% CI. The distribution of CFS was estimated using the Kaplan-Meier method. Logistic regression and Cox proportional hazard models were used to estimate odds ratios (ORs) for best overall response and hazard ratios (HRs) for CFS, respectively. Fisher exact test was used to compare somatic copy-number alterations (SCNAs) and genomic subsets (2-sided). Wilcoxon signed rank test (paired data) and Wilcoxon rank-sum test (independent) were used to analyze both circulating and tissue-based immune profiling parameters (2-sided), using a Bonferroni-Dunn correction for tests of multiple comparisons. Data as of September 30, 2022, were analyzed. Data analysis used R, version 4.3.0 for Windows (R Foundation for Statistical Computing). A 2-sided P ≤ .05 indicates statistical significance.

Results

Between January 10, 2019, and December 13, 2021, the trial enrolled 33 patients. All began protocol treatment and were included in analyses (Figure 1A). Median (range) age was 63 (32-80) years, with a slight majority of female individuals (18 [55%]), and many were smokers (16 [48%]) (Table 1). Eight (24%) had a history of surgically treated early-stage OSCC. Median (range) disease-free interval for those with a head and neck cancer prior to trial entry was 10.5 (0.3-195.0) months. A median of 4 cycles of therapy were received (12% of patients received fewer than all 4 doses).

Twelve patients (36%) (95% CI, 20.4%-54.8%) demonstrated a best overall response of MR or PR, with 3 (9%) demonstrating a greater than 80% reduction in composite score (Table 2). Among individual patients, 2 (6%) had complete resolution of at least 1 target lesion. Sixteen (48%) had SD, and 4 patients (12%) had a best response of PD (Figure 1B and D). Three of the patients with a best response of PD developed OSCC in a target lesion identified on their end-of-treatment biopsy, and the other experienced an increase in the severity of dysplasia in a buccal gingiva target lesion resulting in greater than 20% composite score increase. No patient developed CIS. Six additional patients with a best response other than PD later developed OSCC (eTable 1 in Supplement 2); of note, 6 of the 9 who developed OSCC had a history of early-stage OSCC, and 3 of 12 responders (25%) later developed OSCC. Among the 9 patients with an OSCC event, median (range) time from trial registration to a first OSCC event was 6.6 (1.3-24.3) months, and median time from the last dose of nivolumab to the development of OSCC was 3.7 months. Eight of 9 events were in target lesions.

At a median (range) follow-up of 21.1 (5.4-43.6) months, median CFS had not been reached (NR) (95% CI, 24.3 months to NR) with a 2-year CFS of 72.8% (95% CI, 52.6%-85.5%) (Figure 2). There were 9 CFS events (27.3%) and no deaths (all OSCC events). No clinical or pathologic features appeared to be associated with CFS except a history of early-stage OSCC (HR, 13.53; 95% CI, 3.3-55.5) (eTable 2 in Supplement 2). The median CFS for patients with a prior oral cavity cancer diagnosis was 1.3 months (95% CI, 6.2-12.1), and for patients without a history of OSCC, the median was not reached.

Fatigue was the most common AE (18 [55%]), followed by oral pain (11 [33%]) and diarrhea (9 [27%]) (eTable 3 in Supplement 2). Seven patients (21%) developed grade 3 to 4 AEs, which later resolved. One patient without a cardiac history had atypical chest pain after a half-marathon after cycle 1 and had an elevated troponin T level; cardiologic evaluation clarified a low suspicion for immune-related myocarditis, and the patient resumed treatment. Two patients developed immune-related hepatitis. One patient developed immune-related colitis 5 months after completion of therapy.

All pretreatment dysplastic specimens were evaluable for PD-L1 combined positive score (CPS) testing. Scores ranged from 0 to 80 (eFigure 2A and B in Supplement 2), with 22 (67%) demonstrating a CPS of 1 or greater. No significant difference was observed in PD-L1 CPS scores among responders vs nonresponders (12.5 vs 5, P = .21), and patients with CPS 20 or greater vs less than 20 were not significantly more likely to respond (OR, 4.29; 95% CI, 0.83-25.94) (eTable 4 in Supplement 2).

Multiparametric flow on paired dysplastic tissue before and after treatment revealed that CD8+ T cells showed greater activation (CD69) and immune checkpoint LAG3 coexpression after treatment, with increased LAG3 expression among patients with pretreatment 9p21.3 loss of heterozygosity profiles. Among paired peripheral blood samples, PD-1 expression on both circulating CD4+ and CD8+ T cells decreased significantly (both adjusted P < .001), while CD38 increased on CD8+ T cells (adjusted P < .001) (supporting data in eFigure 2C in Supplement 2).

A subset of 23 patients (70%) had adequate tissue for WES. Twenty pairs of paired peripheral blood and oral dysplastic tissue passed quality controls. Pretreatment median (range) tumor mutational burden was 3.4 (1.4-8.0) mutations per megabase and was similar regardless of response (3.6 vs 2.8, P = .63) and among patients who developed cancer vs not (3.9 vs 2.9, P = .51) (Figure 3A). Genomic driver alterations were similar in patients who developed OSCC vs not. Missense mutations in PIK3CA were common. SCNAs revealed a range of complex allelic-imbalance profiles, primarily focal deletions, most frequently observed at 1q44 (Figure 3B). Only 9p21.3 deletion yielded statistically significant differences between patients who developed OSCC and those who did not. Of 10 patients whose pretreatment tissue sequencing showed 9p21.3 copy-number loss, 6 (60%) later developed OSCC, whereas none of the 10 patients without 9p21.3 loss developed OSCC (P = .01).

Discussion

We present the first (to our knowledge) trial demonstrating the potential efficacy of PD-1 immune checkpoint blockade among patients with high-risk oral precancerous disease. Our data suggest that PD-1 inhibition may yield clinical-pathologic regression in some patients. While some chemoprevention trials have yielded short-term responses to reverse or mute oral carcinogenesis, no therapeutic agents have demonstrated an improvement in CFS, and rates of progression to cancer range from 10% to 30%.79,2528

PVL is an uncommon variant of leukoplakia, occurring in less than 1% of adults, which is aggressive and challenging to treat29,30 largely due to nonhomogeneous, multifocal lesions, and with the histologic hallmarks being corrugated hyperkeratosis and verrucous hyperplasia with variable dysplasia.5,31,32 Some degree of dysplasia was required in our trial with the aim of selecting the highest-risk lesions. Our previous retrospective cohort of patients with PVL suggested a 2-year CFS of 82%.14 In the present trial, we observed a 2-year CFS of 73%; however, we designed the trial with stringent entry criteria, requiring biopsy-proven dysplasia and permitting a history of OSCC. Notably, CFS was a secondary end point in our trial, and the sample size and median follow-up time were limited. It is plausible that our preliminary CFS rate would have been similar without immunotherapy exposure, supporting the need for randomized data. Three patients who had a response during the trial later developed OSCC, suggesting that our scoring system and response definitions may not adequately predict CFS. The prognostic impact of tumor size may not be readily generalizable to precancerous lesions, and a 1-tier change in histopathology (degree of dysplasia) may not be an optimal outcome measurement. As compared to prior chemoprevention trials, our rate of progression to cancer (27%) was comparable,79,2528 while response was defined in prior studies primarily based on lesion size and not histologic change.

Of 9 OSCC events, 6 (67%) were among patients with prior early-stage OSCC with a short median time to failure (<4 months). Including patients with prior cancer events added some heterogeneity to the trial population, but we thought it was important to include them given their recurrence risk.33 Exclusion of patients with prior oral cancer has been implemented in some chemoprevention studies,27,28,34,35 but in the Erlotinib Prevention of Oral Cancer (EPOC) trial,7 60% of patients had prior OSCC. That study followed a prevention-adjuvant therapy convergent design36 under the assumption that high-risk patients with oral premalignant lesions and resected cancers share molecular alterations for prevention and could be studied in similar settings.23,37,38 The cancer events among the patients in the current trial were most often pT1 lesions, but structured follow-up may have identified cancers earlier with a bias toward earlier biopsy. Longer follow-up in a larger randomized trial design will be needed to identify a time-to-event or survival benefit. It is unclear whether immunotherapy favorably affects the pathologic severity of future oral cancer events.

We acknowledge that novel pathologic criteria were required to evaluate clinical activity in this first oral precancer immune checkpoint therapy (ICT) prevention trial, as more traditional response criteria would not apply. We chose a modified composite scoring method to quantify response as a function of lesion size and dysplasia across multiple sites, recognizing that analyzing percent changes in composite score can be limited by small sample size and variability in scores. To limit interobserver variability, we required digital intraoral photography with bidimensional measurements and structured pathologic examination among 2 to 3 oral pathologists. We recognize that distinguishing mild dysplasia from hyperkeratosis can be subject to interpretation, and most of the cohort (73%) had mild dysplasia at baseline. Further, we observed a mix of lesion size and/or histologic changes in response to therapy among individual patients. We appreciate that multifocal lesions may have affected response assessments, but we would not expect spontaneous clinical regression in PVL in the absence of an effective therapy. A time-to-event CFS end point may be more generalizable and have broader clinical applicability. It is also worth noting that the trial population had limited racial and ethnic diversity, and many patients traveled to our center for treatment, which introduced some component of socioeconomic bias. This not only has potential treatment outcome implications, but also may influence tolerance and affordability.

A major concern for this ICT trial was safety, as we treated patients who did not have documented oral cancer. Frequently reported AEs were in line with prior head and neck cancer study populations more broadly.1013 We observed some increase in grade 3 to 4 AEs (21.2%), although we permitted a history of autoimmune disease, and all higher-grade irAEs resolved in time with no deaths. These findings need to be weighed carefully against the potential for clinical activity, given concern for a narrow therapeutic risk-benefit ratio.

Patients with PD-L1 CPS scores of 20 or greater in PVL were not more likely to respond (as has been observed in advanced OSCC11), but this could be due to sample size limitations and/or scoring criteria (CPS is validated on invasive cancers); future studies will need to explore this further. Circulating CD4+/CD8+ T cells displayed significantly increased CD38 and reduced PD-1 expression following treatment, confirming on-target blockade of PD-1 and T-cell activation. An immune phenotype indicative of activation and/or reinvigoration was also observed on CD8+ T cells in oral dysplastic lesions, where surface expression of CD69 and LAG-3 were elevated in posttreatment samples.

Genomic studies of precancers have been limited by adequate tissue availability from small biopsies. Therefore, prior studies generally assessed single genes and/or allelic-imbalance using microsatellite markers, detecting 9p21.3 loss of heterozygosity in approximately 45% of patients with PVL, depending on the number of markers used.39 To our knowledge, this is the first study using WES in PVL, which revealed a range of complex SCNA and allelic-imbalance profiles. Recent data from several groups have found that 9p deletions encompassing 9p21 are significant and selective predictors of ICT resistance in advanced OSCC and lung cancer.23,24,40,41 This may be due to deletions encompassing the type I interferon gene cluster,42 which is often co-deleted with the tumor suppressor CDKN2A, highlighting a key mechanism of immune evasion.43 In our immunogenomic studies, only pretreatment 9p21.3 deletion yielded statistically significant differences: 6 of 10 patients with 9p21.3 deletion in baseline biopsies later developed cancer. We have previously shown that 9p21.3 copy-number loss is generally a focal event in oral precancer44 and associated with an immune-cold signal in OSCC23 that is enhanced by larger deletions extending to the telomeric band at 9p24.1.40 We speculate that resistance to the PD-1 inhibitor in this aggressive oral precancerous disease trial may have arisen during ICT resulting from increasing 9p deletion size to encompass 9p24.1, leading to low expression of the therapeutic target (PD-L1) and other immune gene depletion.23,40,45 PD-L1 is encoded by the CD274 gene, which is located on 9p24.1, close to 9p21, and is often co-deleted in advanced human papillomavirus–negative head and neck squamous cell carcinoma and lung cancers.42,44

Limitations

This study has limitations. We thought it was important to include patients with prior cancer events given their risk but recognize that this added some heterogeneity to the study population. This trial was single group and single center, using a novel clinical and pathologic scoring system to assess immunotherapy response in a hard-to-study oral precancer population. A lack of randomization or use of a time-to-event end point is another limiting factor to acknowledge.

Conclusions

We report the first (to our knowledge) nonrandomized clinical trial of ICT in patients with precancerous disease, specifically patients with high-risk oral precancer, to mitigate progression to OSCC. This trial met its primary response end point, but few patients had complete lesion regression. Other studies using immunotherapy to treat patients with high-risk oral premalignant lesions are ongoing.46,47 Recognizing the limitations and complexity of measuring treatment outcomes in precancer trials, for the first time, we demonstrate potential clinical activity and acceptable safety with the use of ICT in a population with high-risk precancer. A next step would be to consider a larger, precision immunotherapy randomized clinical trial favoring CFS as a primary outcome and stratified by prior history of early-stage treated OSCC and 9p21.3 loss.

Source: JCO

NICE Recommends New Treatment for Advanced Melanoma


The National Institute for Health and Care Excellence (NICE) has recommended the drug combination nivolumab-relatlimab (Opdualag, Bristol Myers Squibb) as a first-line treatment for advanced melanoma. 

This follows licensing by the Medicines and Healthcare Products Regulatory Agency (MHRA) last month as part of Project Orbis, a global partnership with other national regulators to speed review and approval of promising cancer drugs. The drug was approved by the US Food and Drug Administration (FDA) in March 2022 and by the European Commission in September 2022 .

Improved Progression-Free Survival

NICE’s final draft guidance recommended nivolumab-relatlimab for untreated unresectable or metastatic melanoma in people aged 12 years and older, after evidence from the RELATIVITY-047 clinical trial showed that the combined treatment was more effective at slowing progression than nivolumab alone, with a median progression-free survival of 10.1 months compared with 4.6 months, respectively. Progression-free survival at 12 months was 47.7% compared with 36.0%. 

In addition, NICE said, nivolumab-relatlimab was as effective as nivolumab plus ipilimumab, the current standard treatment for advanced melanoma. Indirect comparisons suggested that progression-free survival was also better on nivolumab-relatlimab than on pembrolizumab. There was also indirect evidence suggesting that nivolumab-relatlimab offered an overall survival advantage compared with other treatments. 

Nivolumab and relatlimab are immune checkpoint inhibitors, monoclonal antibodies designed to recognise and attach to specific target proteins: programmed cell death protein 1 (PD1) in the case of nivolumab, and lymphocyte-activation gene 3 (LAG-3) in the case of relatlimab. These proteins switch off T cell activity, so blocking their actions helps to increase T cell activity and aid the immune system response to target and kill melanoma cells.

Melanoma Incidence Rising

According to Cancer Research UK, there are around 16,700 new melanoma cases in the UK every year, making it the fifth most common cancer and accounting for 4% of all new cancer cases. Incidence has risen 140% since the early 1990s, and rates are projected to rise to around 26,500 new cases annually by 2038-2040.

Nivolumab-relatlimab is given by intravenous infusions over 30 minutes every 4 weeks, and treatment must be undertaken in hospital under the supervision of an experienced clinician. NICE recommends the combination treatment be given for 2 years. This reflects clinical practice for these types of drugs where treatment is routinely stopped after this time to reduce the risk of toxicity, the regulator said. 

Adverse Effects Common

Treatment should be stopped earlier if the cancer progresses, and the side effect profile of the combination may also lead to early treatment cessation. Common side effects, most often fatigue, nausea, and diarrhoea, may affect more than one in ten people. Other side effects include myalgia, arthralgia, skin rash, pruritus, anorexia, headache, constipation, vomiting, stomach pain, fever, cough, difficulty breathing, hypothyroidism, vitiligo, and urinary tract infection. 

Serious adverse reactions include adrenal insufficiency, anaemia, colitis, myocarditis, and pneumonia. In the RELATIVITY-047 trial, grade 3 or 4 treatment-related adverse events occurred in 18.9% of patients in the nivolumab-relatlimab group versus 9.7% of patients in the nivolumab group.

NICE estimated that 1293 people could benefit from access to the drug combination, which costs £6135 per 16 mg/mL vial. The recommendation is conditional on the company supplying it at a discount according to a confidential commercial arrangement.

Perioperative Nivolumab and Chemotherapy in Stage III NSCLC


The addition of nivolumab to platinum-based chemotherapy improved pathologic complete response rates in patients with resectable stage IIIA or IIIB NSCLC.

Approximately 20% of patients with non–small-cell lung cancer (NSCLC) have stage III disease. Although therapeutic intent is curative for patients with locally advanced disease, historically, treatment outcomes have been poor, and there is lack of consensus on the most appropriate management.

In the industry-sponsored, open-label, multicenter, phase 2 NADIM II trial, 86 treatment-naive patients with resectable stage IIIA or IIIB NSCLC were randomized 2:1 to receive either neoadjuvant nivolumab and paclitaxel plus carboplatin (experimental group) or paclitaxel plus carboplatin alone (control group), followed by surgery. Patients in the experimental group who had R0 resections received adjuvant nivolumab for 6 months.

Pathologic complete response (pCR), the primary endpoint, occurred in 37% of patients in the experimental group compared with 7% in the control group (relative risk, 5.34; 95% CI 1.34–21.23; P=0.02). Progression-free survival at 24 months was 67.2% in the experimental group and 40.9% in the control group (hazard ratio for disease progression, disease recurrence, or death, 0.47; 95% CI, 0.25–0.88).

During neoadjuvant treatment, grade 3 or 4 adverse events occurred in 19% of patients in the experimental group compared with 10% in the control group, most commonly febrile neutropenia (5%) and diarrhea (4%). A higher percentage of patients in the experimental group underwent surgery (93% vs. 69%); there were no delays in surgery due to adverse events. All patients who attained pCR were free from progression at the time of data cutoff.

Comment

In the NADIM II trial, patients with stage IIIA or stage IIIB NSCLC who were treated with neoadjuvant nivolumab and paclitaxel plus carboplatin achieved a higher rate of pCR and longer survival than those treated with chemotherapy alone. These findings add further support for a neoadjuvant chemo-immunotherapy strategy as demonstrated in CheckMate 816 (NEJM JW Oncol Hematol Apr 14 2022 and N Engl J Med 2022; 386:1973) and in KEYNOTE 677 (NEJM JW Oncol Hematol Jun 20 2023 and N Engl J Med 2023 Jun 3; [e-pub]) in a restricted population of patients with stage III NSCLC.

Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial


Abstract

Background: Patients with metastatic urothelial carcinoma have a dismal prognosis and few treatment options after first-line chemotherapy. Responses to second-line treatment are uncommon. We assessed nivolumab, a fully human IgG4 PD-1 immune checkpoint inhibitor antibody, for safety and activity in patients with metastatic or surgically unresectable urothelial carcinoma whose disease progressed or recurred despite previous treatment with at least one platinum-based chemotherapy regimen.

Methods: In this multicentre, phase 2, single-arm study, patients aged 18 years or older with metastatic or surgically unresectable locally advanced urothelial carcinoma, measurable disease (according to Response Evaluation Criteria In Solid Tumors v1.1), Eastern Cooperative Oncology Group performance statuses of 0 or 1, and available tumour samples for biomarker analysis received nivolumab 3 mg/kg intravenously every 2 weeks until disease progression and clinical deterioration, unacceptable toxicity, or other protocol-defined reasons. The primary endpoint was overall objective response confirmed by blinded independent review committee in all treated patients and by tumour PD-L1 expression (≥5% and ≥1%). This trial is registered with ClinicalTrials.gov, number NCT02387996, and is completed. Follow-up is still ongoing.

Findings: Between March 9, 2015, and Oct 16, 2015, 270 patients from 63 sites in 11 countries received nivolumab, and 265 were evaluated for activity. Median follow-up for overall survival was 7·00 months (IQR 2·96-8·77). Confirmed objective response was achieved in 52 (19·6%, 95% CI 15·0-24·9) of 265 patients. Confirmed objective response was achieved in 23 (28·4%, 95% CI 18·9-39·5) of the 81 patients with PD-L1 expression of 5% or greater, 29 (23·8%, 95% CI 16·5-32·3) of the 122 patients with PD-L1 expression of 1% or greater, and 23 (16·1%, 95% CI 10·5-23·1) of the 143 patients with PD-L1 expression of less than 1%. Grade 3-4 treatment-related adverse events occurred in 48 (18%) of 270 patients-most commonly grade 3 fatigue and diarrhoea, which each occurred in five patients. Three deaths were attributed to treatment (pneumonitis, acute respiratory failure, and cardiovascular failure).

Interpretation: Nivolumab monotherapy provided meaningful clinical benefit, irrespective of PD-L1 expression, and was associated with an acceptable safety profile in previously treated patients with metastatic or surgically unresectable urothelial carcinoma.

Funding: Bristol-Myers Squibb.

Immunotherapy after Surgery Shows Long-Term Benefits for High-Risk Bladder Cancer


Updated results from a large clinical trial confirm that, for some people with bladder cancer that can be removed with surgery, receiving immunotherapy immediately afterwards is an effective treatment.

In 2021, initial results from the same trial led the Food and Drug Administration (FDA) to approve the immune checkpoint inhibitor nivolumab (Opdivo) as a post-surgical (adjuvant) treatment for people with what is called high-risk bladder cancer.

That approval was based on data showing that, compared with treatment with a placebo, giving nivolumab for a year after surgery doubled the amount of time people lived without their disease recurring in or near the bladder or elsewhere in the body, a measure called disease-free survival.

The updated results, presented on February 17 at the 2023 American Society of Clinical Oncology (ASCO) Genitourinary Cancers Symposium, included about 3 years of follow up. These results showed that people who received nivolumab had a median disease-free survival of 22 monthsExit Disclaimer, compared with about 11 months for those who received a placebo.

The picture looked even better for people whose tumors express a protein called PD-L1, which interacts with another protein targeted by nivolumab. That group of patients had a median disease-free survival of more than 52 months.

In the trial, nivolumab was given for a maximum of 1 year, said Matt Galsky, M.D., from the Icahn School of Medicine at Mt. Sinai, who presented the updated results at the ASCO symposium. 

“If you’re just suppressing cancer with a treatment, then potentially, once treatment stops, the cancer starts to grow again,” he said. The long-term survival free of disease seen in some of the study participants “might indicate that [immunotherapy] is actually eradicating the cancer in some patients.”

A lingering risk of recurrence

Bladder cancer that has invaded the bladder muscle or nearby lymph nodes can potentially be cured by surgical removal of the bladder and lymph nodes. But, in more than half of people who have this type of surgery, called radical cystectomy, cancer cells have already spread (metastasized) elsewhere in the body.

These deposits of metastatic cancer “are too small to be seen on [an imaging] scan,” said Dr. Galsky. “But over time those cancer cells grow and divide. And we want to try and avoid that if possible,” he explained.

The main strategy that clinical trials have tested to prevent bladder cancer from recurring, either at or near the original tumor or at distant locations, has been adjuvant therapy: chemotherapy or immunotherapy drugs given after surgery. 

Chemotherapy regimens based on the drug cisplatin are known to shrink tumors in people with bladder cancer that has metastasized at the time it’s diagnosed, explained Andrea Apolo, M.D., of the Genitourinary Malignancies Branch in NCI’s Center for Cancer Research. So these regimens are often used in bladder cancer that can be surgically removed, either before surgery (called neoadjuvant therapy) or afterward, as adjuvant therapy. 

But many people can’t tolerate cisplatin, she added. “And a lot of people prefer not to get chemotherapy because of the side effects,” she said.

Researchers have been searching for other adjuvant therapy options. A previous clinical trial tested a different immunotherapy called atezolizumab (Tecentriq) as an adjuvant therapy, but it did not improve disease-free survival

So for many years, explained Dr. Galsky, most people, following surgery for bladder cancer, underwent observation instead of adjuvant therapy. Observation is following patients with frequent imaging to try to catch a recurrence as early as possible.

But when a previous study found that nivolumab could shrink tumors in people with more advanced bladder cancer, researchers decided to test the drug as an adjuvant treatment in people without detectable metastases.

Preventing disease recurrence for years

The CheckMate 274 trial, which was funded by Bristol Myers Squibb and Ono Pharmaceutical, enrolled more than 700 people with high-risk, muscle-invasive bladder cancer who had undergone extensive surgery. Participants were eligible if they had received neoadjuvant cisplatin-based chemotherapy but not if they had received any adjuvant treatment. 

The researchers randomly assigned trial participants to receive up to a year of treatment with either nivolumab or a placebo. At the time of the ASCO presentation, participants had been followed for a minimum of 31 months. 

The incidence of side effects observed during the longer follow-up period was about the same as that seen during the initial 6 months of treatment. Around 18% of people who received nivolumab had at least one serious side effect, compared with 7% of those who received the placebo. Three of the 353 people who received nivolumab died of side effects attributed to the drug. 

Overall, after a median follow up of 36 months, people who received nivolumab lived about twice as long without their disease progressing than those who received a placebo. 

At the time of the initial publication, there was some evidence that people whose tumors expressed PD-L1 were benefitting more from nivolumab than those whose tumors lacked PD-L1. But it was too early to calculate their median disease-free survival.

With the longer follow up, the researchers found that people whose tumors expressed PD-L1 lived more than twice as long without their disease progressing than the group as a whole.

“These results were impressive but not necessarily unexpected, given what we know about that protein and how [it] might relate to sensitivity to [nivolumab],” Dr. Galsky said. 

However, he added, “for all patients in the study, there was a benefit from [receiving] immunotherapy.” Under FDA’s approval, nivolumab can be used for anyone with high-risk bladder cancer, regardless of whether their tumors express PD-L1.

Pinpointing who needs adjuvant therapy

CheckMate 274 participants will continue to be followed to see if those who received nivolumab live longer, a measure called overall survival

The overall survival data will matter, explained Dr. Apolo. “We know we overtreat a lot of patients with adjuvant therapy,” she said. That is, many people who receive it may have been cured with surgery alone. “But we don’t know yet how to pinpoint who those people are.”

If it turns out that there’s no overall survival improvement from adjuvant immunotherapy, it would make more sense to wait until a cancer recurs or metastasizes to give it, she explained.

A planned NCI-supported trial will soon be looking at whether blood tests for circulating tumor DNA could predict which high-risk patients having surgery for bladder cancer actually need adjuvant therapy and who could safely skip it, Dr. Galsky said.

Although CheckMate 274 didn’t find that treatment with nivolumab decreased participants’ overall quality of life, those who received the drug did experience more side effects and the treatment caused several deaths. “So, if we can identify patients who absolutely don’t need treatment, that certainly makes sense,” said Dr. Galsky.

Other immunotherapy drugs are also being tested as adjuvant treatments for patients with high-risk bladder cancer, Dr. Apolo said. For example, NCI researchers recently finished treating participants in a trial called AMBASSADOR, which is comparing pembrolizumab (Keytruda) with observation after surgery.

In the future, researchers are also interested in testing whether giving nivolumab both before and after surgery can reduce the risk of recurrence even further, Dr. Galsky explained. A recent study found that giving pembrolizumab both before and after surgery reduced the risk of recurrence for some people with melanoma, an aggressive type of skin cancer.

“We’ll see if we see the same thing with bladder cancer,” Dr. Galsky said.

Nivolumab After Selective Internal Radiotherapy for Locally Advanced Liver Cancer


Bruno Sangro, MD, PhD, of Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, and colleagues assessed the safety and efficacy of selective internal radiation therapy (SIRT) combined with nivolumab in patients with unresectable hepatocellular carcinoma. This trial focused specifically on patients who were eligible for transarterial chemoembolization (TACE), and the respective results were published in the Journal for ImmunoTherapy of Cancer.

“[This] trial has shown that the combination of SIRT with SIR-Spheres resin microspheres, followed by nivolumab, was safe and active as first-line therapy of patients with locally advanced hepatocellular carcinoma ineligible for TACE, where SIRT alone has failed to prove superiority over the standard of care,” noted the investigators. “The high disease control rate, prolonged time to [disease] progression, and encouraging overall survival suggest the combination could be an option for this population and should be tested in a phase III controlled trial.”

This phase II, open-label trial enrolled 42 patients with immunotherapy-naive, unresectable hepatocellular carcinoma and preserved liver function. Participants underwent SIRT followed by 240 mg of nivolumab every 2 weeks for up to 24 doses, disease progression, or unacceptable toxicity.

The median follow-up was 22.2 months, and the median time to response was 9 weeks; the median duration of response was 31 weeks. The objective response rate was 41.5%, consisting of 5 complete and 12 partial responses; stable disease was the best overall response in 21 patients. The median time to disease progression was 8.8 months, with 28 patients experiencing disease progression and 27 deaths. The median progression-free survival was 9 months, and the median overall survival was 20.9 months.

Adverse events of grade 3 or 4 were reported in 19% of patients, and serious adverse events affected 26%. Although there were no treatment-related deaths, 27 participants discontinued treatment. Adverse and serious adverse events of any grade were observed in 41 and 21 individuals, respectively; treatment-related events of grade 3 or 4 affected 8 and 5 patients.

Neoadjuvant nivolumab treatment improves survival rates in NSCLC patients


Bottom line:Patients with resectable non-small cell lung cancer (NSCLC) who were treated with neoadjuvant nivolumab had improved five-year recurrence-free and overall survival rates compared with historical outcomes.

Background:NSCLC is the most common type of lung cancer and is a leading cause of cancer-related death worldwide. Despite strides in treating metastatic NSCLC, new treatments for earlier-stage disease have only recently emerged, according to Forde.

Rosner added that there is great interest in optimizing neoadjuvant strategies for earlier-stage NSCLCs that are eligible for surgical resection. Rosner is a medical oncology fellow at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and a member of Forde’s research group.

Forde, Rosner, and colleagues previously reported safety and efficacy results from a phase II clinical trial in which patients with stage I-III resectable NSCLC were treated with two doses of neoadjuvant nivolumab. Major pathological responses were observed in 45 percent of patients, independent of tumor PD-L1 expression, and 73 percent of patients whose tumors were surgically resected were recurrence-free 18 months following surgery.

The latest publication reports the final analyses from this trial, including five-year recurrence-free and overall survival rates for the 20 patients who underwent surgical resection.

“To our knowledge, this is the longest follow-up to date for a PD-1/PD-L1 inhibitor in the neoadjuvant setting for any solid tumor,” said Forde.

Results:Among the 20 patients who underwent surgical resection, 12 patients (60 percent) remained recurrence-free five years after surgery, and 16 patients (80 percent) were alive, exceeding the 36 to 68 percent five-year survival rate historically observed for patients with stage I-III NSCLC, Rosner noted. Forde added that the observed patient outcomes after neoadjuvant nivolumab were better than those historically observed among patients treated with neoadjuvant chemotherapy.

The authors also identified major pathologic response after neoadjuvant nivolumab as a potential predictive biomarker of recurrence-free and overall survival. Of the nine patients who had a major pathological response after neoadjuvant nivolumab, eight were alive and cancer-free five years after treatment. One patient experienced a recurrence within the first 10 months after treatment but has since been disease-free after definitive chemoradiation. The one death in this subgroup was unrelated to cancer.

In contrast, six of the 11 patients who did not have a major pathological response experienced disease recurrence, and three of these patients died due to their cancer. These results indicate that a major pathological response following neoadjuvant nivolumab may be associated with a lower risk of disease recurrence and death, although the authors caution that these results are preliminary and require further validation in larger studies.

Neoadjuvant nivolumab did not lead to surgical delays, and there was only one late-onset immune-related adverse event, which occurred 16 months after nivolumab treatment and was successfully managed, the authors noted.

Author’s comments:“The results from the five-year follow-up analysis indicate that neoadjuvant nivolumab was safe in long-term follow-up and led to encouraging survival in this patient cohort,” said Forde. “The long-term safety and efficacy data from this study provide further support for the use of nivolumab in the neoadjuvant setting.”

Neoadjuvant nivolumab in combination with chemotherapy was approved by the U.S. Food and Drug Administration in March 2022 for the treatment of lung cancer. “Further studies will help us determine whether select patients may benefit from immunotherapy alone,” Forde noted.

“An interesting finding from the analysis was the difference in outcomes between patients with and without a major pathological response,” said Rosner. “Although the sample size was small, the results illustrate the potential power of pathological response as a predictive biomarker.”

Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer


Abstract

Background

In an early-phase study involving patients with advanced non–small-cell lung cancer (NSCLC), the response rate was better with nivolumab plus ipilimumab than with nivolumab monotherapy, particularly among patients with tumors that expressed programmed death ligand 1 (PD-L1). Data are needed to assess the long-term benefit of nivolumab plus ipilimumab in patients with NSCLC.

Methods

In this open-label, phase 3 trial, we randomly assigned patients with stage IV or recurrent NSCLC and a PD-L1 expression level of 1% or more in a 1:1:1 ratio to receive nivolumab plus ipilimumab, nivolumab alone, or chemotherapy. The patients who had a PD-L1 expression level of less than 1% were randomly assigned in a 1:1:1 ratio to receive nivolumab plus ipilimumab, nivolumab plus chemotherapy, or chemotherapy alone. All the patients had received no previous chemotherapy. The primary end point reported here was overall survival with nivolumab plus ipilimumab as compared with chemotherapy in patients with a PD-L1 expression level of 1% or more.

Results

Among the patients with a PD-L1 expression level of 1% or more, the median duration of overall survival was 17.1 months (95% confidence interval [CI], 15.0 to 20.1) with nivolumab plus ipilimumab and 14.9 months (95% CI, 12.7 to 16.7) with chemotherapy (P=0.007), with 2-year overall survival rates of 40.0% and 32.8%, respectively. The median duration of response was 23.2 months with nivolumab plus ipilimumab and 6.2 months with chemotherapy. The overall survival benefit was also observed in patients with a PD-L1 expression level of less than 1%, with a median duration of 17.2 months (95% CI, 12.8 to 22.0) with nivolumab plus ipilimumab and 12.2 months (95% CI, 9.2 to 14.3) with chemotherapy. Among all the patients in the trial, the median duration of overall survival was 17.1 months (95% CI, 15.2 to 19.9) with nivolumab plus ipilimumab and 13.9 months (95% CI, 12.2 to 15.1) with chemotherapy. The percentage of patients with grade 3 or 4 treatment-related adverse events in the overall population was 32.8% with nivolumab plus ipilimumab and 36.0% with chemotherapy.

Conclusions

First-line treatment with nivolumab plus ipilimumab resulted in a longer duration of overall survival than did chemotherapy in patients with NSCLC, independent of the PD-L1 expression level. No new safety concerns emerged with longer follow-up.

Discussion

In this phase 3, randomized trial, we found that patients with advanced NSCLC and a PD-L1 expression level of 1% or more who received nivolumab plus ipilimumab had a significantly longer duration of overall survival than those who received chemotherapy as first-line treatment. At 2 years, the rate of ongoing response was 49% with nivolumab plus ipilimumab, as compared with 11% with chemotherapy. The safety of nivolumab plus ipilimumab has been improved in patients with NSCLC with the use of a lower dose and frequency of administration of ipilimumab, as was suggested in the phase 1 dose-finding study.10

In addition, the duration of overall survival was longer with nivolumab plus ipilimumab than with chemotherapy in all the trial patients, including in those with a PD-L1 expression level of less than 1%, a population for whom anti–PD-1 monotherapy has been insufficient. Although the relative benefit of nivolumab plus ipilimumab, as compared with chemotherapy, was numerically greater in patients with a PD-L1 expression level of less than 1% than in those with a PD-L1 expression level of 1% or more, this result was mostly due to variations between the PD-L1 subgroups in both the median duration of survival and in survival rates in the chemotherapy group. The median duration of overall survival and rates of overall survival at 1 year and 2 years with nivolumab plus ipilimumab were nearly identical in these two PD-L1 subgroups. This result is consistent with previous reports involving patients with melanoma and renal-cell carcinoma, which also showed a benefit for nivolumab plus ipilimumab regardless of PD-L1 level.8,9 The precise underpinnings of the diminished dependence on PD-L1 expression with a combination of PD-1 and CTLA-4 inhibition, as compared with anti–PD-1 monotherapy, are unknown. However, we hypothesize that the differential immune effects of CTLA-4 versus PD-1 inhibition17,18 may be particularly critical in PD-L1–negative tumors for recruiting effective antitumor immunity from the peripheral compartment, which is increasingly recognized as an important mechanism of response to immunotherapy.19-21

Combining nivolumab with ipilimumab has proved to be effective in melanoma and renal-cell carcinoma in previous studies,8,9,22 yet a key question before this trial was whether the addition of CTLA-4 inhibition to PD-1 blockade contributes to benefit in patients with NSCLC. Although this trial was not powered to compare the two regimens, our findings show better efficacy with nivolumab plus ipilimumab than with nivolumab monotherapy within the same trial. In particular, we observed higher rates of complete response and a longer median duration of response (a difference of >7 months) in the patients who received nivolumab plus ipilimumab. In addition, among the patients with a PD-L1 expression level of less than 1%, those who received nivolumab plus ipilimumab had longer overall survival and a longer duration of response (a difference of nearly 10 months) than did those who received nivolumab plus chemotherapy, although this analysis was not part of the statistical testing hierarchy.

Biomarkers for predicting an enhanced benefit for combination immunotherapy relative to chemotherapy remain elusive. The design of this trial was informed by phase 1 and 2 single-group studies of nivolumab plus ipilimumab that showed increased response rates in patients with PD-L1–expressing tumors or a high tumor mutational burden in patients with NSCLC.10,23 However, in this large, randomized study, the survival benefit with nivolumab plus ipilimumab over chemotherapy was ultimately similar in the two main PD-L1 subgroups on the basis of a cutoff of 1% of tumor cells. Moreover, based on emerging data related to the tumor mutational burden as a biomarker, CheckMate 227 was amended to add a primary end point of progression-free survival with nivolumab plus ipilimumab versus chemotherapy in patients with a high tumor mutational burden.11 In the current report, although absolute survival with nivolumab plus ipilimumab was greatest in patients with a high tumor mutational burden, a similar relative benefit of nivolumab plus ipilimumab, as compared with chemotherapy, was seen in patients regardless of tumor mutational burden. The unexpected effect of the tumor mutational burden on the overall survival of patients who received chemotherapy may have contributed to these results. Before we initiated this trial, some24-27 but not all28 studies had shown that survival was not affected by tumor mutational burden with chemotherapy treatment. Further understanding of the role of the tumor mutational burden, if any, as a biomarker is warranted before the integration of this factor into clinical practice.

In the primary analysis from this trial, the median duration of overall survival was significantly longer with nivolumab plus ipilimumab than with chemotherapy among patients with advanced NSCLC who had a PD-L1 expression level of 1% or more. In secondary analyses, the duration of overall survival was also longer with nivolumab plus ipilimumab than with chemotherapy in patients with a PD-L1 expression of less than 1% and in all the trial patients.

Source:NEJM

Anti-PD-1 Drugs Impress as Frontline Therapy for Early, Unfavorable Hodgkin’s


3-year PFS of 99%, OS of 100% with nivolumab-chemo, high response rate with pembrolizumab

Patients with early unfavorable-risk Hodgkin lymphoma had near-perfect 3-year survival outcomes following concurrent or sequential treatment with nivolumab (Opdivo) and chemotherapy, a prospective phase II study showed.

After a median follow-up of 40 months, the 3-year progression-free survival (PFS) rate was 99% and the 3-year overall survival (OS) rate was 100%. No secondary malignancies had been reported, and no new safety signals had emerged. Global quality of life improved or did not deteriorate during follow-up, reported Paul J. Bröckelmann, MD, of the University of Cologne in Germany.

“The efficacy is outstanding in this setting, and especially patients who are given a PR [partial response] do not seem to have disease activity and convert to a CR [complete response] during follow-up,” Bröckelmann said during the American Society of Hematologyopens in a new tab or window annual meeting. “We are happy to report … that most toxicities are temporary and of grade 1 and 2.”

“We need to take all of this together and [ask whether] all of our patients need anti-PD-1 blockade plus a nearly full course of chemo and radiotherapy,” he added. “This is what we want to address in the upcoming trial, looking at individualized immunotherapy in early-stage unfavorable Hodgkin lymphoma.”

A second study involving untreated early-stage unfavorable-risk or advanced classic Hodgkin lymphoma treated with pembrolizumab (Keytruda) and chemotherapy showed a PET-negative (Deauville 1-3) rate of 84% in an analysis of the first 50 patients in an ongoing trial. Treatment-related adverse events (TRAEs) were mostly grade 1/2 and more often related to the chemotherapy, reported Ranjana H. Advani, MD, of Stanford Cancer Center in California.

Bröckelmann reported updated results from the multicenter phase II NIVAHL trialopens in a new tab or window, conducted in Germany. A preliminary reportopens in a new tab or window published 2 years ago showed that all but two of 105 evaluable patients responded to nivolumab administered concurrently or sequentially with AVD (doxorubicin, vinblastine, and dacarbazine) chemotherapy. The update focused on PFS, OS, and toxicity and was published simultaneously in the Journal of Clinical Oncologyopens in a new tab or window.

The trial involved a total of 109 patients with early unfavorable classic Hodgkin lymphoma. The patients were randomly assigned to four cycles of concurrent or sequential nivolumab and AVD, followed by involved-site radiotherapy. The previously reported primary analysis showed that more than 90% of patients in each treatment arm achieved a CR by the end of treatment. Bröckelmann said no additional survival events occurred during follow-up.

The patients randomized to concurrent therapy had a 3-year PFS rate of 100%. The sequential group had a 3-year PFS rate of 98%. Both groups had a 3-year OS rate of 100%.

Minimizing Toxicity

During follow-up, three-fourths of patients had at least one adverse event (AE), most of which were grade 1/2. No patients had grade >1 cardiac events, and 95% of patients had a normal left ventricular ejection fraction. Grade 3 adverse events (AEs) occurred in 9% of patients in both arms combined (no grade 4 AEs). Only 5% of patients required corticosteroids, and a fourth of the patients required long-term non-steroid medication for AEs. At last follow-up visit, no patients required corticosteroids, and 15% remained on non-steroid medications.

Hypothyroidism was the most common grade 2 AE (21%) and also the most common nivolumab-related AE, requiring long-term medication in a majority of cases. Women accounted for almost 90% of the patients who developed hypothyroidism.

The follow-up study to NIVAHL, called INDIEopens in a new tab or window, will use the PD-1 inhibitor tislelizumab, and treatment will be driven by PET imaging. All patients will begin with two doses of tislelizumab. Patients who achieve PET-negative status at the point will receive four additional doses of the PD-1 inhibitor. Those who are not PET negative will receive four doses of tislelizumab plus chemotherapy. Involved-site radiotherapy will be reserved for patients who remain PET positive after chemotherapy.

“We want to try to get rid of most of the chemotherapy in this setting,” Bröckelmann said during the discussion that followed his talk. “We have striking early responses clinically, but also with our correlative studies, to single-agent anti-PD-1, and histologic remission is observed and also reversion of the peripheral immune landscape. We feel confident that two doses of tislelizumab can actually induce some sort of complete remission. This is the rapidly and excellently responding patient population where we can try to de-escalate.”

PET-Adapted Pembrolizumab-Chemo

Advani reported preliminary findings from the KEYNOTE-C11opens in a new tab or window study, which involved a mix of patients with newly diagnosed, early unfavorable, and advanced-stage classic Hodgkin lymphoma. Treatment consisted of pembrolizumab followed by AVD. A prior phase II studyopens in a new tab or window of the regimen showed that the median PFS or OS had yet to be reached after a median follow-up of 33 months. No disease progression or deaths had occurred, and no patient discontinued early because of toxicity.

The ongoing KEYNOTE-C11 is evaluating a PET-adapted regimen of sequential therapy with the PD-1 inhibitor plus chemotherapy, followed by pembrolizumab consolidation in early-stage unfavorable or advanced-stage classic Hodgkin lymphoma. Radiotherapy is omitted in all cases. Investigators have enrolled a total of 146 patients, and Advani presented results from a prespecified interim futility analysis involving the first 50 patients enrolled and treated.

All patients received pembrolizumab and AVD chemotherapy, followed by a PET scan. Patients who were PET negative received additional AVD. PET-positive patients received escalated BEACOPP (bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine [Oncovin], procarbazine, and prednisone) chemotherapy. All patients received pembrolizumab consolidation therapy. The 50 patients included in the interim analysis had completed all chemotherapy, 47 treated only with AVD and three who received BEACOPP.

The 146-patient study population consisted of 62 with early unfavorable disease and 84 with advanced disease, including 32 patients with bulky disease at diagnosis.

Seven of the 50 patients had PET-positive results at the end of treatment, and 42 had negative findings. The remaining patient had discontinued.

During initial pembrolizumab, 80% of the 146 patients experienced AEs: 64% had TRAEs, 12% had grade ≥3 TRAEs, and 5% discontinued because of TRAEs. Among 116 patients who received AVD, 76% had AEs of any grade, 64% had TRAEs, 54% had grade ≥3 TRAEs, and no patients discontinued.

The most common pembrolizumab-related AEs were pyrexia, pruritus, elevated alanine transaminase levels, and hyperthyroidism. Most of the AEs were grade 1/2. The most common AEs during AVD were nausea, decreased neutrophil count, and neutropenia.

“Mid-treatment, this PET-adapted regimen showed promising antitumor activity and no safety concerns,” said Advani. “Per study design, patients are continuing with planned therapy. Pembrolizumab induction followed by chemotherapy may be an effective treatment option in patients with newly diagnosed, early unfavorable, or advanced-stage classic Hodgkin lymphoma.”

Souce: Medscape