Abstract
Background
Patients with non‐small‐cell lung cancer (NSCLC) and uncommon EGFR alterations typically have worse treatment outcomes than patients with classically EGFR‐mutated NSCLC. This study aimed to investigate the efficacy and safety of PD‐1 blockade with sintilimab plus anti‐angiogenic treatment with anlotinib in patients with NSCLC harboring uncommon EGFR mutations.
Methods
Patients with metastatic NSCLC harboring uncommon EGFR mutations after two previous treatments, including a platinum‐based chemotherapy regimen and a targeted treatment (or chemotherapy only for patients harboring EGFR ex20ins), received sintilimab combined with anlotinib. The primary endpoint was objective response rate (ORR).
Results
At data cutoff (September 27, 2022), median follow‐up was 22.3 months (range, 1.2–37.6). Among 21 enrolled patients, 12 had EGFR ex20ins and nine had other uncommon EGFR mutations such as L861Q, G719A, and G709X. Overall, eight patients (38.1%) achieved an objective response, and 18 (85.7%) achieved disease control. Median (95% CI) progression‐free survival (PFS) was 7.0 (5.4–8.6) months, and median overall survival (OS) was 20.0 (15.6–24.4) months. The 12‐month PFS rate (95% CI) was 22.2% (7.4–42.0), and the 12‐month OS rate was 66.7% (42.5–82.5). Patients harboring EGFR ex20ins had similar ORR and PFS to those with other mutations. Six patients (28.6%) experienced grade 3 treatment‐related adverse events (TRAEs); hand‐foot syndrome was the most common grade 3 TRAE (2 patients; 9.5%). No grade ≥4 TRAEs were observed.
Conclusions
The combination of sintilimab and anlotinib demonstrated durable efficacy and was generally well tolerated in patients with NSCLC and uncommon EGFR mutations who had received prior standard‐of‐care treatments. (ClinicalTrials.gov identifier: NCT04790409).
Keywords: anlotinib, anti‐angiogenic, non‐small cell lung cancer (NSCLC), PD‐1, sintilimab, uncommon EGFR mutations
1. BACKGROUND
Lung cancer is the leading cause of cancer‐related death globally. 1 Epidermal growth factor receptor (EGFR) gene mutations are the most prevalent oncogenic driver alteration among Asian patients with lung adenocarcinoma and may be present in up to 51.4% of patients. 2 Approximately 90% of EGFR mutations in non‐small cell lung cancer (NSCLC) are “classic mutations” (19del, L858R, etc.) that are sensitive to treatment with EGFR tyrosine kinase inhibitors (TKIs). 3 , 4 Indeed, EGFR TKIs are the recommended first‐line treatment for EGFR‐mutated NSCLC, with an associated median progression‐free survival (PFS) of 9.0–18.9 months. 5 , 6 , 7 , 8 , 9 The other 10% of EGFR mutations in NSCLC comprise rare mutations such as EGFR ex20ins, G719X, L861Q, and S768I/V, which confer inferior responses to EGFR TKI treatment. 2
The EGFR TKIs afatinib and osimertinib are recommended for the treatment of NSCLC harboring G719X, S768I, and L861Q mutations. These agents have demonstrated moderate anti‐tumor activity, with a median PFS of 8.2–10.7 months. 10 , 11 However, when TKI resistance occurs, the only remaining treatment option is platinum‐based chemotherapy, which is associated with a median PFS of only 4–5 months. 12 In addition, patients with less common EGFR mutations typically achieve a poor response to EGFR TKIs. For example, the ex20ins mutation has long been considered an “undruggable target” and is associated with a worse median overall survival (OS) than other uncommon EGFR point mutations. 10 , 13 In one study of patients with ex20ins treated with afatinib, the objective response rate (ORR) was only 8.7%, and the median PFS was 2.7 months, 10 suggesting primary resistance to first‐ or second‐generation EGFR TKIs may be common. To date, amivantamab and mobocertinib (TAK‐788) are approved for the second‐line treatment of patients with NSCLC and ex20ins, with ORRs of 40%, 14 and 28%, 15 respectively. However, as these drugs are not available in many countries, the treatment of patients with ex20ins is modeled on that used for patients with oncogene‐negative NSCLC (e.g., salvage chemotherapy). After progression on first‐line chemotherapy, second‐line docetaxel has limited therapeutic effect and substantial toxicity, with a median OS of only 6 months. 16 Collectively, the limited accessibility and efficacy of targeted therapies for patients with uncommon EGFR mutations (especially ex20ins) mean that new treatment strategies to improve clinical outcomes are urgently needed.
Immune checkpoint inhibitors (ICIs) targeting the programmed death 1 (PD‐1)/PD ligand 1 (PD‐L1) pathway have revolutionized the treatment of advanced NSCLC; however, the observed OS benefit with second‐line ICI monotherapy over docetaxel in patients with NSCLC has failed to extend to EGFR‐mutated patients. 17 , 18 Retrospective studies have suggested that some EGFR mutation subtypes may be associated with sensitivity to ICIs; specifically, patients with ex20ins or G719X appear to achieve a longer PFS and a higher ORR than patients with classic EGFR mutations. 19 , 20
Combining ICIs with antiangiogenic agents can result in synergistic antitumor effects. 21 For example, in the IMpower150 trial, the addition of atezolizumab to bevacizumab and chemotherapy in patients with EGFR‐TKI drug‐resistant NSCLC led to improved efficacy. 22 The Orient‐31 study reported an advantage of a chemotherapy‐containing four‐drug therapy (sintilimab plus bevacizumab biosimilar plus cisplatin and pemetrexed) over chemotherapy alone in patients with EGFR‐mutated NSCLC after TKI failure. 23 However, as these two trials included few patients with non‐classic EGFR mutations (26.0% and 6.3%, respectively) and reported a relatively high proportion of patients experiencing grade 3 or 4 treatment‐related adverse events (TRAEs) with the four‐drug regimen, it is necessary to identify regimens with better tolerability and comparable efficacy for patients with NSCLC harboring rare EGFR mutations.
Sintilimab is a novel anti–PD‐1 monoclonal antibody (mAb) with encouraging antitumor activity in advanced NSCLC. 24 The combination of sintilimab with chemotherapy as first‐line therapy for advanced NSCLC has been shown to improve ORR and PFS. 25 , 26 Anlotinib is an anti‐angiogenic TKI that targets VEGFR 1/2/3, FGFR1‐4, PDGFR a/b, and c‐Kit and has been approved as a third‐line treatment for NSCLC in China. 27 Sintilimab plus anlotinib has shown improved efficacy compared with chemotherapy alone in first‐line for patients with advanced NSCLC. 28 In order to reduce the toxicity associated with the four‐drug regimen and treat NSCLC with rare EGFR mutations after TKI failure, sintilimab plus anlotinib represents a promising treatment choice after failure of standard of care treatment. In this prospective phase 2 study, we investigated the efficacy and safety of sintilimab plus anlotinib without chemotherapy in pretreated patients with advanced NSCLC harboring uncommon EGFR mutations.
2. METHODS
2.1. Study design and participants
This was a single‐center, open‐label, phase II trial conducted at Zhejiang Cancer Hospital, China (ClinicalTrials.gov identifier: NCT04790409). The study was conducted following the provisions of the Declaration of Helsinki and Good Clinical Practice guidelines. The study protocol was approved by the Ethics Commission of Zhejiang Cancer Hospital (IRB‐2019‐81) and written informed consent was provided by all patients before inclusion.
Eligible patients were 18–70 years old and had histologically or cytologically confirmed treatment‐experienced advanced NSCLC (AJCC 8th edition), measurable disease, and disease progression after at least two treatment regimens that included one platinum‐containing chemotherapy and one EGFR TKI. Patients with EGFR ex20ins mutations who had experienced disease progression after only platinum‐based chemotherapy were also eligible. Other key eligibility criteria included an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and adequate organ function. Key exclusion criteria included components of small cell carcinoma, symptomatic central nervous system metastases, central squamous cell carcinoma with a cavity, active hemorrhage or risk of hemorrhage, and prior immunotherapy.
2.2. Systemic therapy
Eligible patients received sintilimab (Innovent [Suzhou] Biopharmaceutical Co., Ltd.) 200 mg intravenously once on Day 1 and subsequently every 3 weeks, plus anlotinib (Chia Tai Tianqing Pharmaceutical Group Co., Ltd.) 12 mg orally on Days 1–14 of the 21‐day treatment cycle, until disease progression or death, unacceptable toxicity, withdrawal of consent, or for a maximum of 24 months (35 cycles) of sintilimab. No other antitumor therapy was allowed before disease progression. For sintilimab, no dose adjustment was allowed. For anlotinib, the investigator could reduce the dose to 10 mg, 8 mg, or permanently discontinue treatment according to drug‐related toxicity. Patients with intolerable adverse events (AEs) that led to a delay or discontinuation of one study drug continued treatment with the other drug.
2.3. Outcomes and measurements
The primary endpoint was ORR, assessed by the investigator according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. Secondary endpoints were PFS, OS, disease control rate (DCR, defined as the best overall response [BOR] of stable disease [SD] or better), duration of response (DOR, defined as the time from first documented complete or partial response [CR or PR] to disease progression or death), and safety and tolerability. Tumor responses were measured using computed tomography or magnetic resonance imaging. Patients underwent imaging evaluations at baseline (treatment initiation), Week 6, Week 12, and then every 9 weeks. After Week 48, imaging evaluations were performed every 12 weeks. Imaging evaluations could be performed within ±7 days of the scheduled visit. CR and PR were confirmed radiologically at least 4 weeks after the first response. Safety was recorded from enrollment until 90 days after the last dose of study treatment. Treatment‐emergent adverse events (TEAEs), treatment‐related adverse events (TRAEs), and laboratory biochemical data parameters were summarized using the Common Terminology Criteria for Adverse Events (CTCAE), version 5.01.
2.4. Biomarker analysis
Before treatment initiation, tumor tissue samples from initial diagnosis or post‐resistance biopsy were collected for evaluation of PD‐L1, CD4, CD8, CD163, Foxp3, and LAG3 expression in the tumor microenvironment. PD‐L1 expression was measured by a 22C3 pharmDx assay (Agilent Technologies), and PD‐L1 positivity (PD‐L1+) was defined as a PD‐L1 tumor proportion score (TPS) ≥1%. CD4, CD8, CD163, Foxp3, and LAG3 staining on immune cells was reported as the proportion of positive cells among all nucleated cells in the stromal compartments, and scoring was recorded as negative (<10%) or positive (≥10%).
Epidermal growth factor receptor mutations were assessed by using amplification refractory mutation system polymerase chain reaction (ARMS‐PCR; Amoy Diagnostics) or next‐generation sequencing (NGS, Burning Rock Biotech and Nanjing Geneseeq Technology Inc.). Among them, 12 patients were detected by ARMS‐PCR and nine patients by NGS. The details of targeted NGS data are described in the Appendix S1. The sequencing coverage and quality statistics for each sample are summarized in Table S1.
2.5. Statistical analysis
The sample size was calculated using a Simon's 2‐stage design 29 with alpha set at 0.1, power set to 0.83 and assuming an ORR of 10% for historical treatment (single‐drug docetaxel monotherapy in second line), and 30% for sintilimab plus anlotinib. Consequently, it was required that 14 eligible patients receive treatment in the first stage of the study with at least one response to continue the enrollment. In stage 2, four additional patients would be enrolled for a total target enrollment of 18. Overall, if four responses or more were observed, the treatment regimen would be considered successful. Allowing for an estimated drop‐out rate of 15%, a sample size of 21 patients was planned.
The main analysis included all patients who met the inclusion criteria and received at least one dose of sintilimab combined with anlotinib (intent‐to‐treat [ITT] population). The safety analysis was also performed in the ITT population. ORR and DCR were analyzed using the Clopper–Pearson method. PFS, OS, and DOR were estimated using the Kaplan–Meier method. Baseline demographics and safety data were summarized using descriptive statistics. Cox proportional‐hazards regression models were used for a univariate analysis to identify potential factors associated with clinical outcomes. All statistical tests were two‐sided, and p < 0.05 was considered statistically significant. All statistical analyses were performed using R software (version 3.4.3).
3. RESULTS
3.1. Patients
Between August 2019 and December 2021, a total of 21 patients were enrolled. Baseline characteristics of the enrolled patients are shown in Table 1. Overall, 66.7% were male, the median age was 61 years, 90.5% had lesions classified histologically as adenocarcinoma, the majority (66.7%) of patients had an ECOG performance status of 1, and 61.9% had never smoked. Prior to enrollment, 15 patients (71.4%) had received ≥2 treatments. Preexisting brain or liver metastases were present in 9 (42.9%) and 4 (19.0%) patients, respectively. Twelve patients (57.1%) had confirmed ex20ins and nine (42.9%) had G719A, L861Q, or G709X EGFR mutations. Seven patients (33.3%) had detectable intra‐tumoral expression of PD‐L1 (TPS ≥1%), while 14 patients (66.7%) had a PD‐L1 TPS that was missing or <1%. Among the 15 patients with evaluable samples, positive expression of CD4+, CD8+, Foxp3+, CD163+, and LAG3+ immune cells was identified in 53.3% (8/15), 33.3% (5/15), 60.0% (9/15), 40.0% (6/15), and 20.0% (3/15), respectively. Dual‐positivity for expression of PD‐L1 and CD8 was identified in 20.0% (3/15) of patients.
TABLE 1.
Patient demographics and baseline characteristics.
| Characteristic | Patients (N = 21) |
|---|---|
| Median (range) age, years | 61 (36–71) |
| Sex | |
| Male | 14 (66.7) |
| Female | 7 (33.3) |
| ECOG performance status | |
| 0 | 7 (33.3) |
| 1 | 14 (66.7) |
| Histology | |
| Adenocarcinoma | 19 (90.5) |
| Non‐adenocarcinoma | 2 (9.5) |
| Smoking history | |
| Never | 13 (61.9) |
| Current/previous | 8 (38.1) |
| Family history of NSCLC | |
| No | 14 (66.7) |
| Yes | 7 (33.3) |
| Uncommon mutation type | |
| G719X, etc. | 9 (42.9) |
| ex20ins | 12 (57.1) |
| Brain metastases | 9 (42.9) |
| Liver metastases | 4 (19.0) |
| Prior lines of treatment | |
| 1 | 6 (28.6) |
| 2 | 11 (52.4) |
| ≥3 | 4 (19.0) |
| PD‐L1 TPS, % | |
| <1 | 8 (38.1) |
| 1–49 | 4 (19.0) |
| >50 | 3 (14.3) |
| Missing | 6 (28.6) |
Note: Data are n (%) unless otherwise stated.
Abbreviations: ECOG, Eastern Cooperative Oncology Group; NSCLC, non‐small cell lung cancer; TPS, tumor proportion score.
At data cutoff (September 27, 2022), the median duration of follow‐up was 22.3 months (range: 1.2–37.6), at which time 10 patients were still alive (three still on treatment), and 11 had died.
3.2. Efficacy
In the ITT population, the majority of patients (61.9%) had a reduction in tumor burden (Figure 1A). Responses were observed as early as 6 weeks (Figure 1B), with eight patients (38.1%) achieving an objective response and 18 (85.7%) with disease control (Table S2). The median DOR was 5.2 months (95% CI: 0.0–11.2). Overall, SD and/or a PR were noted in most patients during the first 6–7 months of combined sintilimab and anlotinib treatment (Figure 2). The ORR was consistent across different EGFR mutation patterns and the presence or absence of liver or brain metastases (EGFR ex20ins, 41.7% vs other mutations, 33.3%; liver metastasis or not: 25.0% vs 41.2%; brain metastasis or not: 33.3% vs 41.7%; all p > 0.05, Table S2).
FIGURE 1.

(A) Best change from baseline in sum of target lesion diameters, according to type of EGFR mutation. Column labels indicate BOR. BOR, best overall response; ex, exon; ins, insertion; PD, progressive disease; PR, partial response; SD, stable disease; (B) Duration of treatment and time to response. BOR, best overall response; PD, progressive disease; PR, partial response; SD, stable disease.
FIGURE 2.

Change from baseline in sum of target lesion diameters over time. Three patients had a BOR of PD; two patients had a change from baseline in target lesion diameters of 24% over 6 weeks, so these data points overlap on the plot. BOR, best overall response; PD, progressive disease; PR, partial response; SD, stable disease.
The median PFS was 7.0 months (95% CI, 5.4–8.6), with a 6‐month PFS rate of 57.1% (95% CI, 33.8–74.9) and a 12‐month PFS rate of 22.2% (95% CI, 7.4–42.0; Figure 3A). The median OS was 20.0 months (95% CI, 15.6–24.4), with a 6‐month OS rate of 71.4% (95% CI, 47.2–86.0) and a 12‐month OS rate of 66.7% (95% CI, 42.5–82.5; Figure 3B). Patients carrying EGFR ex20ins had a similar PFS to patients with other mutation patterns (4.3 vs 7.3 months, p = 0.685). Moreover, patients with brain metastases had a similar PFS to those without brain metastases (6.9 vs 7.9 months, p = 0.254; Figure 3C). Although patients with liver metastases had worse PFS than those without liver metastases, the difference was not statistically significant (1.2 vs 7.0 months, p = 0.944; Figure 3D).
FIGURE 3.

Progression‐free survival (PFS) overall and overall survival (OS) overall and in subgroups of interest. Kaplan–Meier plots indicating: (A) PFS among all patients; (B) OS among all patients; (C) PFS for patients with (red trace) or without (blue trace) brain metastasis; (D), PFS for patients with (red trace) or without (blue trace) liver metastasis.
A univariate analyses showed no statistically significant association between PFS and PD‐L1 expression or the presence of CD4+, CD8+, Foxp3+, CD163+, and LAG3+ immune cells (Figure 4A). There was a trend for patients who were double positive for PD‐L1 and CD8 to have a lower risk for disease progression than those who were not double positive (HR, 0.15 [95% CI, 0.02–1.23]; p = 0.077; Figure 4A) but this result did not reach statistical significance. No factors were observed to be associated with OS (Figure 4B).
FIGURE 4.

(A) Univariate analysis of progression‐free survival according to baseline patient characteristics; (B) Univariate analysis of overall survival according to baseline patient characteristics.
3.3. Safety
The median duration of treatment was 7.0 months (range, 0.7–15.2) for combination therapy. At least one TRAE of any grade occurred in 20 patients (95.2%), the most common of which were hypothyroidism (eight patients, 38.1%), increased aspartate aminotransferase (AST; seven patients, 33.3%) and proteinuria (seven patients, 33.3%; Table 2). Grade 3 TRAEs occurred in six patients (28.6%). The most common grade 3 TRAEs were hand‐foot syndrome (two patients, 9.5%), hypertension (one patient, 4.8%), cerebral infarction (one patient, 4.8%), fatigue (one patient, 4.8%), and immune‐related pneumonitis (one patient, 4.8%; Table 2). Four patients (19.0%) required a dose reduction of anlotinib owing to unacceptable toxicity. Two patients (9.5%) discontinued treatment because of TRAEs (one experienced pneumonitis [anlotinib continued], and one experienced cerebral infarction [discontinued both drugs]). There were no grade 4–5 TRAEs and no new safety signals were observed.
TABLE 2.
Summary of treatment‐related adverse events (intent‐to‐treat population; N = 21).
| TRAE | Any grade | Grade 3 |
|---|---|---|
| Hypothyroidism | 8 (38.1) | 0 |
| Increased AST | 7 (33.3) | 0 |
| Proteinuria | 7 (33.3) | 0 |
| Fatigue | 6 (28.6) | 1 (4.8) |
| Increased ALT | 6 (28.6) | 0 |
| Thrombocytopenia | 6 (28.6) | 0 |
| Hand‐foot syndrome | 5 (23.8) | 2 (9.5) |
| Hyperthyroidism | 4 (19.0) | 0 |
| Rash | 4 (19.0) | 0 |
| Hypertension | 3 (14.3) | 1 (4.8) |
| Anemia | 2 (9.5) | 0 |
| Diarrhea | 2 (9.5) | 0 |
| Hypertriglyceridemia | 2 (9.5) | 0 |
| Hypoalbuminemia | 2 (9.5) | 0 |
| Hyponatremia | 2 (9.5) | 0 |
| Immune‐related pneumonitis | 2 (9.5) | 1 (4.8) |
| Increased creatinine | 2 (9.5) | 0 |
| Increased uric acid | 2 (9.5) | 0 |
| Neutropenia | 2 (9.5) | 0 |
| Cerebral infarction | 1 (4.8) | 1 (4.8) |
| Epistaxis | 1 (4.8) | 0 |
| Hoarseness | 1 (4.8) | 0 |
| Hypercholesterolemia | 1 (4.8) | 0 |
| Increased direct bilirubin | 1 (4.8) | 0 |
| Oral ulcer | 1 (4.8) | 0 |
| Nausea | 1 (4.8) | 0 |
| Vomiting | 1 (4.8) | 0 |
Note: Data are n (%).
Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; TRAE, treatment‐related adverse event.
4. DISCUSSION
To our knowledge, this is the first study to report the efficacy and safety of sintilimab plus anlotinib in pretreated patients with advanced NSCLC harboring uncommon EGFR mutations. Our study revealed that this combined therapy has favorable efficacy, durability, and tolerability. Interestingly, favorable clinical outcomes were observed for patients with PD‐L1 and CD8 dual positivity. Based on our findings, sintilimab plus anlotinib has promise as a chemotherapy‐free treatment regimen for patients with NSCLC harboring uncommon EGFR mutations after disease progression on standard of care treatment.
Previous studies in NSCLC have revealed the effectiveness of combined treatment with ICIs and antiangiogenic therapies. The inhibition of angiogenesis potentiates PD‐1/PD‐L1 blockade by reducing hypoxia, promoting the infiltration of CD8 T cells, and inhibiting recruitment of tumor‐associated macrophages. 30 In the phase 1 JVDF trial, a combination of the anti‐VEGFR‐2 antibody, ramucirumab, plus the anti‐PD‐1 antibody pembrolizumab led to an ORR of 42.3%, including patients previously treated for NSCLC. 31 A phase 1b study of first‐line sintilimab plus anlotinib in patients with advanced NSCLC reported a high ORR (72.7%) and DCR (100%), 32 leading to an ongoing phase 2 randomized study of sintilimab plus anlotinib versus chemotherapy that has so far reported an ORR of 50% (chemotherapy: 33%) and a median PFS of 10.8 months (chemotherapy: 5.7 months; HR = 0.4, p = 0.002). 28 In addition, second‐line therapy with the anti‐PD‐1 mAb camrelizumab and the VEGFR‐2 TKI apatinib in patients with advanced nonsquamous NSCLC was associated with an ORR of 30.9% and a median PFS of 5.7 months. 33
In the current study, 38.1% of patients who received late‐line treatment with sintilimab plus anlotinib achieved an objective response, and 85.7% achieved disease control. Overall, the median PFS was 7.0 months, and median OS was 20.0 months. Interestingly, given the diverse clinical settings, the ORR for sintilimab plus anlotinib observed in the present study (38.1%) compares favorably with previous studies of anlotinib monotherapy (9.2%) 27 and sintilimab monotherapy (25.5%) in NSCLC. 34 A retrospective study of 28 patients harboring EGFR ex20ins who received a PD‐1/PD‐L1 inhibitor (with or without a CTLA‐4 inhibitor), reported an ORR of 10.7% (3/28) and a median PFS of 1.9 months 19 ; seven patients with EGFR G719X who received PD‐1 inhibitor monotherapy had an ORR of 28.6% and a PFS of 4.8 months. 19 However, cabozantinib plus atezolizumab only demonstrated modest activity in 30 patients with previously‐treated advanced EGFR‐mutated NSCLC (ORR = 7%, DCR = 63%, median PFS = 2.7 months, and median OS = 6.1 months). 35 In contrast, anlotinib combined with ICIs exhibited a longer median PFS (4.3 months vs 3.6 months, p = 0.005), and OS (14.2 months vs 9.0 months p = 0.029) than chemotherapy for the late‐line treatment of patients with NSCLC and EGFR‐TKI resistance. 36 Additionally, the phase 3 Orient‐31 trial reported that sintilimab plus the bevacizumab biosimilar IBI305 and chemotherapy led to significant improvements in PFS over chemotherapy alone in patients with advanced EGFR‐mutated NSCLC after TKI failure (6.9 vs 4.3 months; p < 0.001). 37 ORR was also higher in the four‐drug group versus chemotherapy (44% vs 25%). 37 Taken together, these data show that sintilimab plus anlotinib has exhibited better anti‐tumor activity than other similar combinations of a multi‐target angiogenesis inhibitor and ICI and shown comparable efficacy to a four‐drug regimen in patients with pre‐treated NSCLC with uncommon EGFR mutations.
While anti‐tumor activity was observed across multiple subgroups in this study, we noted a trend towards higher ORR for patients harboring EGFR ex20ins versus other EGFR tumor mutations (41.7% vs 33.3%). However, the statistical analysis was limited by the small sample size. The development of selective TKIs (e.g., mobocertinib, sunvozertinib, and CLN‐081) and a bi‐specific antibody (amivantamab) represent major advances for patients with this hard‐to‐treat molecular alteration. However, ORRs and median PFS values are yet to exceed 41% and 12 months, respectively. 38 , 39 , 40 Therefore, new treatment strategies, especially those containing ICIs, are necessary in patients with NSCLC harboring EGFR ex20ins. In this regard, the combination of sintilimab and anlotinib is a promising treatment regimen in this patient population.
Among the nine patients with brain metastases at baseline, those with brain metastases had a similar overall PFS to those without brain metastases. Although only two patients had evaluable brain lesions, one achieved intracranial PR and one achieved intracranial SD. Both patients sustained intracranial benefit when extracranial lesions showed progression. The intracranial responses observed were active, although this requires further validation in larger patient cohorts.
In this study, we evaluated the association between survival outcomes and tumor PD‐L1 expression and immune‐cell infiltration. Our results suggest that PFS and OS following treatment with sintilimab combined with anlotinib are not affected by PD‐L1 status, in line with prior studies of ICIs combined with antiangiogenic agents in patients with metastatic NSCLC. 30 , 32 PD‐L1 expression can be induced by genetic alterations and inflammatory cytokines, such as interferon (IFN)‐γ. However, in EGFR‐mutated tumors, PD‐L1 expression may only reflect EGFR signaling rather than effector T‐cell activity, 41 , 42 abrogating the predictive utility of PD‐L1 expression in EGFR‐mutated NSCLC. In the present study, the efficacy of combination therapy was similar between patients with different abundances of immune cells (CD4+, CD8+, Foxp3+, CD163+, and LAG3+). It is possible that these single biomarkers do not comprehensively reflect the characteristics of the tumor microenvironment in EGFR‐mutated NSCLC, resulting in poor predictive performance. 43 Additionally, consistent with our preclinical research, 44 patients with dual positivity for PD‐L1 and CD8 TILs showed a trend toward a lower risk of disease progression versus those without dual positivity (HR, 0.15 [95% CI, 0.02–1.23]), although this result did not reach statistical significance and the statistical analysis was limited by the small sample size. To date, data on the mechanism and predictive role of PD‐L1 expression and immune‐cell infiltration in NSCLC with uncommon EGFR mutations are scarce, and further research is needed to inform patient stratification based on these biomarkers.
Combined treatment with sintilimab plus anlotinib was generally well tolerated, and the safety profile was consistent with that of either drug as monotherapy. 27 , 45 The incidence of severe grade 3 TRAEs (28.6%) was relatively low, and there were few treatment discontinuations due to TRAEs. Hand‐foot syndrome was the most common grade ≥3 TRAE (9.5%), which is consistent with the known side effects of angiogenesis inhibitors. 46 No grade 4 or 5 TRAEs were observed.
Limitations of this study include its single‐arm design with a relatively small sample size and a potential for selection bias due to the lack of a control group. In addition, immunohistochemistry results could not be obtained from every patient due to sample accessibility, and there were variable tumor tissue resources. Therefore, the observed trend toward better response among patients with dual expression of PD‐L1/CD8 requires further prospective research for verification.
5. CONCLUSIONS
This phase 2 study demonstrated that the combination of sintilimab and anlotinib has promising anti‐tumor activity and is generally well tolerated in patients with advanced, pre‐treated NSCLC harboring uncommon EGFR mutations. Further evaluation of this treatment strategy in a well‐designed, prospective trial with a larger cohort is warranted.
AUTHOR CONTRIBUTIONS
Kaiyan Chen: Conceptualization (lead); funding acquisition (supporting); writing – original draft (lead). Yanjun Xu: Data curation (equal); formal analysis (equal). Zhiyu Huang: Data curation (equal). Xiaoqing Yu: Data curation (equal). Wei Hong: Data curation (equal). Hui Li: Formal analysis (equal). Xiaoling Xu: Formal analysis (equal). Hongyang Lu: Data curation (equal). Fajun Xie: Formal analysis (equal). Jun Chen: Formal analysis (equal). Youzu Xu: Formal analysis (equal). Yun Fan: Funding acquisition (lead); project administration (lead); supervision (lead); writing – review and editing (lead).
FUNDING INFORMATION
This work was supported by Beijing Xisike Clinical Oncology Research Foundation (Grant Y‐XD2019‐052), Medical Health Science and Technology Project of Zhejiang Province (Grant 2023KY601), and National Natural Science Foundation of China (Grant 81972718).
CONFLICT OF INTEREST STATEMENT
None of the authors has a conflict of interest related to the study.
ETHICS STATEMENT
This study conformed to the provisions of the Declaration of Helsinki and Good Clinical Practice guidelines. The study protocol was approved by the Ethics Commission of Zhejiang Cancer Hospital (IRB‐2019‐81) and written informed consent was provided by all patients before inclusion.
Supporting information
Appendix S1.
ACKNOWLEDGMENTS
We thank the patients and the families of the patients who participated in this study.
Chen K, Xu Y, Huang Z, et al. Sintilimab plus anlotinib as second‐ or third‐line therapy in metastatic non‐small cell lung cancer with uncommon epidermal growth factor receptor mutations: A prospective, single‐arm, phase II trial. Cancer Med. 2023;12:19460‐19470. doi: 10.1002/cam4.6548
Kaiyan Chen and Yanjun Xu contributed equally to this work.
DATA AVAILABILITY STATEMENT
The data underlying this article will be shared on reasonable request to the corresponding author.
REFERENCES
- 1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7‐33. [DOI] [PubMed] [Google Scholar]
- 2. Tan AC, Tan DSW. Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. J Clin Oncol. 2022;40(6):611‐625. [DOI] [PubMed] [Google Scholar]
- 3. D'Angelo SP, Pietanza MC, Johnson ML, et al. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol. 2011;29(15):2066‐2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Rosell R, Moran T, Queralt C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361(10):958‐967. [DOI] [PubMed] [Google Scholar]
- 5. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open‐label, first‐line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non‐small‐cell lung cancer in Asia (IPASS). J Clin Oncol. 2011;29(21):2866‐2874. [DOI] [PubMed] [Google Scholar]
- 6. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first‐line treatment for European patients with advanced EGFR mutation‐positive non‐small‐cell lung cancer (EURTAC): a multicentre, open‐label, randomised phase 3 trial. Lancet Oncol. 2012;13(3):239‐246. [DOI] [PubMed] [Google Scholar]
- 7. Park K, Tan EH, O'Byrne K, et al. Afatinib versus gefitinib as first‐line treatment of patients with EGFR mutation‐positive non‐small‐cell lung cancer (LUX‐Lung 7): a phase 2B, open‐label, randomised controlled trial. Lancet Oncol. 2016;17(5):577‐589. [DOI] [PubMed] [Google Scholar]
- 8. Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first‐line treatment for patients with EGFR‐mutation‐positive non‐small‐cell lung cancer (ARCHER 1050): a randomised, open‐label, phase 3 trial. Lancet Oncol. 2017;18(11):1454‐1466. [DOI] [PubMed] [Google Scholar]
- 9. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR‐mutated advanced non‐small‐cell lung cancer. N Engl J Med. 2018;378(2):113‐125. [DOI] [PubMed] [Google Scholar]
- 10. Yang JC, Sequist LV, Geater SL, et al. Clinical activity of afatinib in patients with advanced non‐small‐cell lung cancer harbouring uncommon EGFR mutations: a combined post‐hoc analysis of LUX‐Lung 2, LUX‐Lung 3, and LUX‐Lung 6. Lancet Oncol. 2015;16(7):830‐838. [DOI] [PubMed] [Google Scholar]
- 11. Cho JH, Lim SH, An HJ, et al. Osimertinib for patients with non‐small‐cell lung cancer harboring uncommon EGFR mutations: a multicenter, open‐label, phase II trial (KCSG‐LU15‐09). J Clin Oncol. 2020;38(5):488‐495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Mok TSK, Kim SW, Wu YL, et al. Gefitinib plus chemotherapy versus chemotherapy in epidermal growth factor receptor mutation‐positive non‐small‐cell lung cancer resistant to first‐line gefitinib (IMPRESS): overall survival and biomarker analyses. J Clin Oncol. 2017;35(36):4027‐4034. [DOI] [PubMed] [Google Scholar]
- 13. Zhang T, Wan B, Zhao Y, et al. Treatment of uncommon EGFR mutations in non‐small cell lung cancer: new evidence and treatment. Transl Lung Cancer Res. 2019;8(3):302‐316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Park K, Haura EB, Leighl NB, et al. Amivantamab in EGFR exon 20 insertion‐mutated non‐small‐cell lung cancer progressing on platinum chemotherapy: initial results from the CHRYSALIS phase I study. J Clin Oncol. 2021;39(30):3391‐3402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Duke ES, Stapleford L, Drezner N, et al. FDA approval summary: mobocertinib for metastatic non‐small cell lung Cancer with EGFR Exon 20 insertion mutations. Clin Cancer Res. 2022;29(3):508‐512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hanna N, Shepherd FA, Fossella FV, et al. Randomized phase III trial of pemetrexed versus docetaxel in patients with non‐small‐cell lung cancer previously treated with chemotherapy. J Clin Oncol. 2004;22(9):1589‐1597. [DOI] [PubMed] [Google Scholar]
- 17. Lee CK, Man J, Lord S, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non‐small cell lung carcinoma: a systematic review and meta‐analysis. JAMA Oncol. 2018;4(2):210‐216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. 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(2):403‐407. [DOI] [PubMed] [Google Scholar]
- 19. Hastings K, Yu HA, Wei W, et al. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non‐small‐cell lung cancer. Ann Oncol. 2019;30(8):1311‐1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Chen Q, Shang X, Liu N, et al. Features of patients with advanced EGFR‐mutated non‐small cell lung cancer benefiting from immune checkpoint inhibitors. Front Immunol. 2022;13:931718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti‐angiogenic treatments and vice versa. Nat Rev Clin Oncol. 2018;15(5):310‐324. [DOI] [PubMed] [Google Scholar]
- 22. Reck M, Mok TSK, Nishio M, et al. Atezolizumab plus bevacizumab and chemotherapy in non‐small‐cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open‐label phase 3 trial. Lancet Respir Med. 2019;7(5):387‐401. [DOI] [PubMed] [Google Scholar]
- 23. Lu S, Wu L, Jian H, et al. Sintilimab plus chemotherapy for patients with EGFR‐mutated non‐squamous non‐small‐cell lung cancer with disease progression after EGFR tyrosine‐kinase inhibitor therapy (ORIENT‐31): second interim analysis from a double‐blind, randomised, placebo‐controlled, phase 3 trial. Lancet Respir Med. 2023;11(7):624‐636. [DOI] [PubMed] [Google Scholar]
- 24. Hoy SM. Sintilimab: first global approval. Drugs. 2019;79(3):341‐346. [DOI] [PubMed] [Google Scholar]
- 25. Zhou C, Wu L, Fan Y, et al. Sintilimab plus platinum and gemcitabine as first‐line treatment for advanced or metastatic squamous NSCLC: results from a randomized, double‐blind, phase 3 trial (ORIENT‐12). J Thorac Oncol. 2021;16(9):1501‐1511. [DOI] [PubMed] [Google Scholar]
- 26. Yang Y, Sun J, Wang Z, et al. Updated overall survival data and predictive biomarkers of Sintilimab plus pemetrexed and platinum as first‐line treatment for locally advanced or metastatic nonsquamous NSCLC in the phase 3 ORIENT‐11 study. J Thorac Oncol. 2021;16(12):2109‐2120. [DOI] [PubMed] [Google Scholar]
- 27. Han B, Li K, Wang Q, et al. Effect of anlotinib as a third‐line or further treatment on overall survival of patients with advanced non‐small cell lung cancer: the ALTER 0303 phase 3 randomized clinical trial. JAMA Oncol. 2018;4(11):1569‐1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Han B, Chu T, Yu Z, et al. LBA57 Sintilimab plus anlotinib versus platinum‐based chemotherapy as first‐line therapy in metastatic NSCLC (SUNRISE): an open label, multi‐center, randomized, phase II study. Ann Oncol. 2022;33:S1423‐S1424. [Google Scholar]
- 29. Jung SH, Lee T, Kim K, George SL. Admissible two‐stage designs for phase II cancer clinical trials. Stat Med. 2004;23(4):561‐569. [DOI] [PubMed] [Google Scholar]
- 30. Zhao S, Ren S, Jiang T, et al. Low‐dose apatinib optimizes tumor microenvironment and potentiates antitumor effect of PD‐1/PD‐L1 blockade in lung cancer. Cancer Immunol Res. 2019;7(4):630‐643. [DOI] [PubMed] [Google Scholar]
- 31. Herbst RS, Arkenau HT, Santana‐Davila R, et al. Ramucirumab plus pembrolizumab in patients with previously treated advanced non‐small‐cell lung cancer, gastro‐oesophageal cancer, or urothelial carcinomas (JVDF): a multicohort, non‐randomised, open‐label, phase 1a/b trial. Lancet Oncol. 2019;20(8):1109‐1123. [DOI] [PubMed] [Google Scholar]
- 32. Chu T, Zhong R, Zhong H, et al. Phase 1b study of sintilimab plus anlotinib as first‐line therapy in patients with advanced NSCLC. J Throrac Oncol. 2021;16(4):643‐652. [DOI] [PubMed] [Google Scholar]
- 33. Zhou C, Wang Y, Zhao J, et al. Efficacy and biomarker analysis of camrelizumab in combination with apatinib in patients with advanced nonsquamous NSCLC previously treated with chemotherapy. Clin Cancer Res. 2021;27(5):1296‐1304. [DOI] [PubMed] [Google Scholar]
- 34. Shi Y, Wu L, Yu X, et al. Abstract CT041: ORIENT‐3: a randomized, open‐label, phase 3 study of sintilimab versus docetaxel in previously treated advanced/metastatic squamous non‐small‐cell lung cancer (sqNSCLC). Cancer Res. 2021;81(13_Supplement):CT041‐CT041. [Google Scholar]
- 35. Neal JW, Lim FL, Aix SP, et al. EP08.02‐081 cabozantinib plus atezolizumab in first or second‐line advanced NSCLC and previously‐treated EGFR mutant advanced NSCLC. J Thorac Oncol. 2022) S439;17(9):S439. [Google Scholar]
- 36. Yu L, Hu Y, Xu J, et al. Multi‐target angiogenesis inhibitor combined with PD‐1 inhibitors may benefit advanced non‐small cell lung cancer patients in late line after failure of EGFR‐TKI therapy. Int J Cancer. 2023;153(3):635‐643. [DOI] [PubMed] [Google Scholar]
- 37. Lu S, Wu L, Jian H, et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR‐mutated non‐squamous non‐small‐cell lung cancer who progressed on EGFR tyrosine‐kinase inhibitor therapy (ORIENT‐31): first interim results from a randomised, double‐blind, multicentre, phase 3 trial. Lancet Oncol. 2022;23(9):1167‐1179. [DOI] [PubMed] [Google Scholar]
- 38. Zhou C, Ramalingam SS, Kim TM, et al. Treatment outcomes and safety of mobocertinib in platinum‐pretreated patients with EGFR exon 20 insertion‐positive metastatic non‐small cell lung cancer: a phase 1/2 open‐label nonrandomized clinical trial. JAMA Oncol. 2021;7(12):e214761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Riely GJ, Neal JW, Camidge DR, et al. Activity and safety of mobocertinib (TAK‐788) in previously treated non‐small cell lung cancer with EGFR exon 20 insertion mutations from a phase I/II trial. Cancer Discov. 2021;11(7):1688‐1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Passiglia F, Malapelle U, Normanno N, Pinto C. Optimizing diagnosis and treatment of EGFR exon 20 insertions mutant NSCLC. Cancer Treat Rev. 2022;109:102438. [DOI] [PubMed] [Google Scholar]
- 41. Sugiyama E, Togashi Y, Takeuchi Y, et al. Blockade of EGFR improves responsiveness to PD‐1 blockade in EGFR‐mutated non‐small cell lung cancer. Sci Immunol. 2020;5(43):eaav3937. [DOI] [PubMed] [Google Scholar]
- 42. Dong ZY, Zhang JT, Liu SY, et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD‐1 blockade in non‐small cell lung cancer. Onco Targets Ther. 2017;6(11):e1356145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Qiao M, Jiang T, Liu X, et al. Immune checkpoint inhibitors in EGFR‐mutated NSCLC: dusk or dawn? J Thorac Oncol. 2021;16(8):1267‐1288. [DOI] [PubMed] [Google Scholar]
- 44. Chen K, Cheng G, Zhang F, et al. PD‐L1 expression and T cells infiltration in patients with uncommon EGFR‐mutant non‐small cell lung cancer and the response to immunotherapy. Lung Cancer. 2020;142:98‐105. [DOI] [PubMed] [Google Scholar]
- 45. Gao S, Li N, Gao S, et al. Neoadjuvant PD‐1 inhibitor (Sintilimab) in NSCLC. J Thorac Oncol. 2020;15(5):816‐826. [DOI] [PubMed] [Google Scholar]
- 46. Ding F, Liu B, Wang Y. Risk of hand‐foot skin reaction associated with vascular endothelial growth factor‐tyrosine kinase inhibitors: a meta‐analysis of 57 randomized controlled trials involving 24,956 patients. J Am Acad Dermatol. 2020;83(3):788‐796. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Appendix S1.
Data Availability Statement
The data underlying this article will be shared on reasonable request to the corresponding author.
