ArticlePDF Available

Phase 1b Study of Sintilimab Plus Anlotinib as First-line Therapy in Patients With Advanced NSCLC

Authors:
  • School of Mechanics and Aerospace Engineering Southwest Jiaotong University

Abstract and Figures

Introduction: Although the interaction between tumor immune microenvironment and angiogenesis has been well established, evidence supporting the chemo-free combination of immune checkpoint inhibitors plus antiangiogenic TKIs in treatment naïve advanced NSCLC patients is insufficient. This report provides the efficacy and safety of sintilimab combined with anlotinib as first-line therapy for advanced NSCLC from a phase Ib trial (NCT03628521). Methods: Eligible patients, who were treatment-naïve and had unresectable stage IIIB/C or IV NSCLC without EGFR/ALK/ROS1 mutations, received sintilimab (200mg day1) and anlotinib (12mg day1-14) q3w till disease progression or unacceptable toxicity. Baseline PD-L1 expression and tumor mutation burden status were assessed in all patients. The primary endpoints were objective response rate and safety. Results: Twenty-two patients received sintilimab and anlotinib. Median follow-up was 15.8 months (range, 8.3-19.3). 16 patients achieved confirmed partial response, the ORR was 72.7% (95% CI, 49.8%-89.3%), and DCR was 100% (95% CI, 84.6%-100%). Median PFS was 15 months (95% CI, 8.3m, NR), and the 12-month PFS rate was 71.4% (95% CI, 47.2%-86.0%). The incidence rate of grade ≥3 TRAEs was 54.5%, and grade 3 hypertension was predominant (2/22, 9.1%). No grade 4 TRAEs were observed, and one case of grade 5 immune-related pneumonitis occurred. Conclusions: To the best of our knowledge, this is the first study that has assessed an anti-PD-1 antibody combined with a multi-target antiangiogenic TKI in the front-line setting for NSCLC patients. In view of its encouraging efficacy, durability, and safety profile, sintilimab plus anlotinib represents a novel chemotherapy-free regimen in this patient population.
Content may be subject to copyright.
ORIGINAL ARTICLE
Phase 1b Study of Sintilimab Plus Anlotinib as
First-line Therapy in Patients With Advanced
NSCLC
Tianqing Chu, MD,
a
Runbo Zhong, MD,
a
Hua Zhong, MD,
a
Bo Zhang, MD,
a
Wei Zhang, MD,
a
Chunlei Shi, MD,
a
Jialin Qian, MD,
a
Yanwei Zhang, MD,
a
Qing Chang, MD,
a
Xueyan Zhang, MD,
a
Yu Dong, MD,
a
Jiajun Teng, MD,
a
Zhiqiang Gao, MD,
a
Huiping Qiang,
a
Wei Nie, MD,
a
Yiming Zhao, PhD,
a
Yuchen Han, PhD,
b
Ya Chen,
a
Baohui Han, MD
a,
*
a
Respiratory Department, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, Peoples Republic of China
b
Pathology Department, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, Peoples Republic of China
Received 20 October 2020; revised 15 November 2020; accepted 20 November 2020
Available online - 29 January 2021
ABSTRACT
Introduction: Although the interaction between tumor
immune microenvironment and angiogenesis has been well
established, evidence supporting the chemo-free combina-
tion of immune checkpoint inhibitors plus antiangiogenic
tyrosine kinase inhibitors in treatment-naive patients with
advanced NSCLC is insufcient. This report provides the
efcacy and safety of sintilimab combined with anlotinib as
rst-line therapy for advanced NSCLC from a phase 1b trial
(NCT03628521).
Methods: Eligible patients who were treatment-naive and
had unresectable stage IIIB/C or IV NSCLC without EGFR/
ALK/ROS1 mutations received sintilimab (200 mg, day 1)
and anlotinib (12 mg, day 114) every 3 weeks till disease
progression or unacceptable toxicity. Baseline programmed
death-ligand 1 expression and tumor mutation burden
status was assessed in all patients. The primary end points
were objective response rate and safety.
Results: A total of 22 patients received sintilimab and
anlotinib. Median follow-up was 15.8 months (range: 8.3
19.3). Sixteen patients achieved conrmed partial response
with an objective response rate of 72.7% (95% condence
interval [CI]: 49.8%89.3%) and disease control rate of
100% (95% CI: 84.6%100%). Median progression-free
survival was 15 months (95% CI: 8.3 m, not reached), and
the 12-month progression-free survival rate was 71.4%
(95% CI: 47.2%86.0%). The incidence rate of grade 3 or
higher treatment-related adverse events was 54.5%, and
grade 3 hypertension was predominant (two of 22, 9.1%).
No grade 4 treatment-related adverse events were
observed, and one case of grade 5 immune-related pneu-
monitis occurred.
Conclusions: To the best of our knowledge, this is the rst
study that assessed an antiprogrammed cell death protein
1 antibody combined with a multitarget antiangiogenic
tyrosine kinase inhibitor in the frontline setting for patients
with NSCLC. In view of its encouraging efcacy, durability,
and safety prole, sintilimab plus anlotinib represents a
novel chemotherapy-free regimen in this patient population.
2020 Published by Elsevier Inc. on behalf of International
Association for the Study of Lung Cancer.
Keywords: Nonsmall cell lung cancer; First-line; AntiPD-1;
Antiangiogenic TKIs; Chemotherapy-free
Introduction
Immune checkpoint inhibitors (ICIs) have emerged in
rst- and second-line therapy for NSCLC and become a
dynamic eld in which new combinations are constantly
being evaluated. Whereas antiangiogenic therapy has
exhibited the potential to directly or indirectly alleviate
immunosuppression,
1
conversely, ICIs can prompt tu-
mor vessel normalization.
2
This suggests that immuno-
therapy and antiangiogenesis may have synergistic
*Corresponding author.
Disclosure: The authors declare no conict of interest.
Address for correspondence: Baohui Han, MD, Respiratory Department,
Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, Peo-
ples Republic of China. E-mail: 18930858216@163.com
ª2020 Published by Elsevier Inc. on behalf of International
Association for the Study of Lung Cancer.
ISSN: 1556-0864
https://doi.org/10.1016/j.jtho.2020.11.026
Journal of Thoracic Oncology Vol. 16 No. 4: 643-652
antitumor effects. The clinical benets of combining ICIs
and antiangiogenic agents have been reported in a va-
riety of clinical studies of solid tumors.
3-7
In particular,
the IMpower150 trial has reported an enhanced efcacy
of atezolizumab and bevacizumab in combination with
chemotherapy in chemotherapy-naive patients, making
this regimen one of the current standard of care treat-
ments for advanced nonsquamous NSCLC.
8
Despite the
observed efcacy in the IMpower150 study, this
chemotherapy-containing four-drug regimen led to a
high incidence of grade 3 or 4 treatment-related adverse
events (TRAEs).
8
Given that severe AEs may deteriorate
treatment compliance, performance status and quality of
life, and even subsequent treatment eligibility, there is
an unmet need for a more tolerable regimen with com-
parable efcacy for patients with NSCLC. Meanwhile,
some nonchemotherapy trials have found good tolera-
bility and efcacy across multiple tumor types, which
suggest the worth of investigating ICIs plus anti-
angiogenic agents in the frontline setting for advanced
NSCLC.
3-7,9
Sintilimab is a fully-humanized immunoglobulin G4
antiprogrammed cell death protein 1 (PD-1) mono-
clonal antibody with encouraging antitumor activity in
advanced NSCLC.
9-12
Anlotinib blocks tumor angiogen-
esis and proliferation by inhibiting the vascular endo-
thelial growth factor (VEGF) receptors (1/2/3) and other
major tyrosine kinase receptors, such as FGFR1-4,
PDGFR a/b, c-Kit, and FLT3.
13
In phase 3 ALTER 0303
trial, anlotinib substantially prolonged progression-free
survival (PFS) and overall survival (OS) in patients
with advanced NSCLC who failed at least second-line
therapy.
14
On the basis of such favorable results, we
conducted this phase 1b study to evaluate the efcacy
and safety of the combination of sintilimab and anlotinib
as a chemotherapy-free regimen in the rst-line treat-
ment for patients with advanced NSCLC.
Materials and Methods
Design and Patients
This was a single-center, open-label, phase 1b trial
conducted in Shanghai Chest Hospital. It was a part of
the NCT03628521 trial (cohort C), which evaluated
anlotinib-based combination therapy as a rst-line
treatment for advanced NSCLC. The results of each
cohort of the study will be reported separately. The
study was conducted in accordance with the Declaration
of Helsinki and Good Clinical Practice guidelines. The
study protocol was reviewed and approved by the
institutional review board and ethics committee at
Shanghai Chest Hospital. All patients provided written
informed consent before enrollment.
Eligible patients had the following inclusion criteria:
(1) 18 to 75 years of age; (2) histologically conrmed
NSCLC at the clinical stage of IIIB/IIIC (inoperable or not
suitable for denitive concurrent chemoradiotherapy) or
stage IV, as dened by American Joint Committee on
Cancer, eighth edition; (3) without EGFR/ALK/ROS1
mutations; (4) Eastern Cooperative Oncology Group
performance status score of 0 to 1; and (5) no previous
systemic therapy for advanced diseases. The main
exclusion criteria were the following: (1) SCLC
(including patients with mixed small cell and non-small
cell tumors); (2) symptomatic or had uncontrolled brain
metastases (BMs); (3) central squamous cell carcinoma
with a cavity; (4) and with active hemorrhage or at the
risk of hemorrhage.
Treatment
Sintilimab (200 mg, Innovent [Suzhou] Biopharma-
ceutical Co., Ltd.) was intravenously administered on day
1 of 3-week cycles, and anlotinib (12 mg, Chia Tai
Tianqing Pharmaceutical Group Co., Ltd.) was orally
administered once daily on day 1 to 14 per cycle. The
combination of sintilimab plus anlotinib would be
assessed for safety in 10 patients in the rst cycle before
accruing the additional patients targeted for study entry.
If any of those 10 patients experienced intolerable AEs,
which were dened as grade 3 hematologic toxicity for
more than 7 days, grade 4 hematologic toxicity, or higher
than grade 3 nonhematologic toxicity, patients subse-
quently enrolled would receive a reduced dose of anlo-
tinib at 10 mg or 8 mg. Treatment continued until
disease progression, intolerance, or patientswithdrawal
of consent. No other antitumor therapy was allowed
before disease progression. Patients with intolerable AEs
that lead to delay or discontinuation of one drug
continued the treatment with the other study drug.
Biomarker Analysis
Before the treatment, the patientstumor tissue
samples would be collected for programmed death-
ligand 1 (PD-L1) expression evaluation and tumor mu-
tation burden (TMB) assessment. PD-L1 expression was
measured by 22C3 pharmDx assay (Agilent Technolo-
gies, Carpinteria, CA), and PD-L1positive (PD-L1
þ
) was
dened as PD-L1 tumor proportion score greater than or
equal to 1%. The TMB was measured by the Founda-
tionOne CDx assay (Foundation Medicine, Cambridge,
MA), and TMB-high (TMB-H) was dened as greater than
or equal to 10 mutations per Mb. Blood samples would
be collected at baseline, before and after the treatment
for subsequent exploratory studies, such as T-cell re-
ceptor dynamics during the treatment.
644 Chu et al Journal of Thoracic Oncology Vol. 16 No. 4
Assessment
Tumor response was assessed by investigators per
Response Evaluation Criteria in Solid Tumors 1.1 every 6
weeks (±7 d). Complete response (CR) or partial
response (PR) would be conrmed in subsequent
radiographic assessments at least 4 weeks later. Patients
with rst radiologic evidence of progressive disease (PD)
would continue on treatment until disease progression
was conrmed in subsequent examinations provided
that they would benet from continuous treatment. All
patients underwent brain magnetic resonance imaging at
baseline, and for those with BMs, the intracranial
response would be evaluated per Response Evaluation
Criteria in Solid Tumors 1.1 concurrently at scheduled
tumor assessments. All patients would be followed up
every 2 months for survival after PD.
Safety was assessed throughout the study. AEs were
graded according to the U.S. National Cancer Institutes
Common Terminology Criteria for Adverse Events
(version 4.0) and recorded from enrollment until 90
days after the last dose of study drugs.
End Point
The primary end points were objective response rate
(ORR) (dened as the proportion of patients with CR
plus PR) and safety. The secondary end points included
disease control rate (DCR) (dened as the proportion of
patients with CR, PR, and stable disease maintained 6
wk), duration of response (DOR), PFS, and OS. DOR was
dened as the time from rst documented CR or PR to
disease progression or death. PFS was dened as the
time from the rst dose of either drug to investigator-
assessed radiologic PD or death from any cause and
would be censored at the time of last tumor assessment.
OS was dened as the time from patient enrollment to
death from any cause.
Statistics
Efcacy and safety were evaluated in all patients who
received at least one dose of the study drugs. The data
were analyzed by descriptive statistical analyses. Contin-
uous data were expressed as mean (±SD) or median with
an interquartile range as appropriate. Categorical data
were expressed in numbers and percentages. ORR and
DCR were calculated with corresponding two-sided 95%
condence intervals (CIs) using the Clopper-Pearson
method. The Kaplan-Meier method was used for the
analysis of PFS and OS. All data analysis was done with
Statistical Analysis System version 9.4 software.
Results
Patient Enrollment and Baseline Characteristics
Between September 2018 and February 2019, a
total of 24 patients were enrolled. Among them, 22
received at least one dose of study treatment
(Supplementary Fig. 1) and were included in the ef-
cacy and safety analysis. Two were excluded before
the treatment owing to the lack of measurable lesions
and inadequate biopsy tissue. Patients were predomi-
nately men (95.5%) and with an Eastern Cooperative
Oncology Group performance status score of 1
(95.5%). A total of 13 patients (59.1%) had stage IV
disease, and patients at stage IIIB/IIIC were all N3
with supraclavicular lymph node metastases. A total of
12 (54.5%) had squamous cell carcinomas, nine
(40.1%) had adenocarcinomas, and one was diagnosed
as not otherwise specied. Among the 13 stage IV
patients, four had BM, and two had liver metastasis at
baseline. PD-L1 expression and TMB status were
assessed in all 22 patients. Because of tissue sample
disqualication, three were not evaluable for TMB, and
one was not assessable for neither TMB nor PD-L1.
Among the patients with evaluable biomarkers, 13
(59.1%) were PD-L1
þ
, and seven (31.8%) were TMB-
H. The detailed baseline characteristics of the partici-
pants are illustrated in Table 1. At the time of data
cutoff (April 30, 2020), the median follow-up time was
15.8 months (range: 8.319.3), and nine patients were
still under the study of treatment. The treatments
paused in 10 patients during the lockdown for coro-
navirus disease 2019, and the median pause time was
3 weeks (range: 26). All patients resumed the treat-
ment after being radiologically conrmed as progres-
sion-free.
Efcacy
Out of 22 patients, 16 achieved conrmed PR, and the
ORR was 72.7% (Table 2). DCR was 100%, and all pa-
tients achieved tumor shrinkage (Fig. 1A). The median
time to response was 1.6 months (95% CI: 1.42.9). The
responses were durable, with a median DOR not reached
(NR) (95% CI: 3.2NR) (Table 2 and Fig. 1B). Objective
responses were noted in eight of nine patients (88.9%)
with adenocarcinoma and seven of 12 (58.3%) patients
with squamous cell carcinomas. Patients had a consis-
tent ORR cross the stage subgroups (stage IIIB/IIIC, six
of nine, 66.7% versus stage IV, 10 of 13, 76.9%). The
benet of sintilimab plus anlotinib combination, in terms
of ORR, was regardless of PD-L1 expression level (PD-
L1
þ
versus PD-L1
, 69.2% versus 75.0%) or TMB
(TMB-H versus TMB-low, 85.7% versus 63.6%). Among
the four patients with BM, three achieved overall PR, and
the intracranial responses were all CR. Till the last follow-
up, two PR patients had sustained intracranial and sys-
temic responses. One PR patient had disease progression
at primary lesion after 6.7 months. The patient who
achieved stable disease had disease progression at both
intracranial and primary sites after 4.2 months. Till the
April 2021 Sintilimab Plus Anlotinib in First Line NSCLC 645
cutoff date, no newly developed brain metastases were
observed in patients without BM at baseline.
PFS events occurred in nine patients (40.9%), and
the 12-month PFS rate was 71.4% (95% CI: 47.2%
86.0%). The median PFS was 15 months (95% CI: 8.3
moNR) (Fig. 2 and Supplementary Table 1). In sub-
group analyses, the 12-month PFS rates were similar
between the patients with squamous cell carcinoma and
those with adenocarcinoma (72.7% versus 66.7%). The
median PFS was 13 months in the squamous cell car-
cinoma subgroup, whereas it was NR in the adenocar-
cinoma subgroup. Patients at stage IIIB/IIIC had a
similar median PFS with stage IV (15 versus 14 mo). The
12-month PFS rate of patients without BM was 76.5%
(95% CI: 48.8%90.4%), and the median PFS was 15
months. The 12-month PFS rate of patients with BM was
50.0% (95% CI: 5.78%84.5%), whereas the median
PFS was NR (Supplementary Fig. 2 and Supplementary
Table 1). Compared with PD-L1
patients, PD-L1
þ
pa-
tients had a longer median PFS (NR versus 14 mo) and a
higher 12-month PFS rate (76.9% versus 62.5%). No
disease progression was observed in all seven TMB-H
patients until the end of the follow-up period, and the
12-months PFS rate was 60.0% for TMB-low patients.
OS data was immature owing to the limited occurrence
of death (n ¼3) at the time of data cutoff, and the
estimated 12-month OS rate was 95.5% (95% CI:
71.9%99.3%).
Figure 1. Best overall response and drug exposure. (A) Best overall response. (B) Duration of drug exposure. BM, brain
metastases; EOT, end of treatment; NA, not applicable; PD, progressive disease; PD-L1, programmed death-ligand 1; PR,
partial response; TMB, tumor mutation burden.
646 Chu et al Journal of Thoracic Oncology Vol. 16 No. 4
Safety
The median treatment duration was 14.6 months
(range: 3.719.3) for combination therapy, 14.1 months
(range: 2.919.3) for sintilimab, and 14.0 months (range:
3.719.3) for anlotinib, respectively. No AEs higher than
grade 2 were observed in the rst 10 patients during the
rst cycle; therefore, all enrolled patients started with
anlotinib at the dose of 12 mg. Five (22.7%) patients
required a dose reduction of anlotinib (n ¼410 mg and
n¼18 mg) owing to intolerable toxicities during the
study period. One patient discontinued anlotinib (owing
to grade 3 cerebral infarction), one discontinued sintili-
mab (owing to grade 1 immune-mediated pneumonitis),
and one discontinued both drugs (owing to grade 3
rash).
TRAEs occurred in all patients, and the most common
TRAEs were hemorrhage (59.1%), hypothyroidism
(50.0%), and increased uric acid (40.9%) (Table 3).
Grade 3 TRAEs occurred in 12 patients (54.5%). Except
for two cases of hypertension (Table 3), most of the
grade 3 TRAEs occurred only once.
The most common antiangiogenetic therapyrelated
AEs were hemorrhage (59.1%), hypertension (31.8%),
and proteinuria (18.2%). Among the 84.6% (11 of 13)
patients, hemorrhage events were grade 1, with three
patients experiencing transient grade 1 hemoptysis. Only
one patient with grade 2 urinary occult blood required
medication. No higher than grade 3 hemorrhage
occurred, and no treatment was interrupted or dis-
continued owing to hemorrhage events. The incidence
rate of hemorrhage was similar between patients with
squamous cell carcinoma and adenocarcinoma (66.7%
versus 55.6%). The most common immune-related AE
were hypothyroidism (50%), pneumonitis (13.6%),
myositis (4.5%), and adrenal insufciency (4.5%). Only
one patient experienced both grade 3 myositis and ad-
renal insufciency. No grade 4 AEs were observed.
One patient with grade 5 immune-related AE was
recorded, who had developed grade 2 immune-related
pneumonitis after eight cycles of treatment and
improved after intravenous methylprednisolone and
anti-infective medication. The patient died 2 months
later from unexplained acute respiratory failure. How-
ever, the radiographic assessment did not reveal subse-
quent pneumonitis deterioration; therefore, it was
unlikely that the cause of death was associated with
immune-related pneumonitis.
Discussion
In this phase 1b trial, sintilimab plus anlotinib as the
rst-line therapy has exhibited favorable efcacy, dura-
bility, and tolerability in treatment-naive patients with
advanced NSCLC without sensitive mutations. The clin-
ical benets were consistent across different stage, his-
tologic, or molecular subgroups. It is noteworthy that,
despite the different clinical settings, patients receiving
this combined regimen had a highly superior ORR
(72.7%) compared with anlotinib (9.2%) or sintilimab
(20%) alone.
12,14
Besides, such response was compara-
ble to that of sintilimab-based chemotherapy in rst-line
advanced or metastatic NSCLC (68.4% and 64.7% in
nonsquamous and squamous, respectively) in a phase 1b
study.
9
Thus, sintilimab plus anlotinib could represent a
promising chemotherapy-free option for treatment-naive
patients with advanced NSCLC.
There have been several promising clinical outcomes
with ICIs plus antiangiogenic antibodies in advanced
NSCLC
15
; however, more effective combinations remain
to be developed. In the phase I JVDF trial, the combina-
tion of pembrolizumab and ramucirumab exhibited an
ORR of 42.3% and a 12-month PFS rate of 45% in
Table 1. Baseline Patient Characteristics
Characteristics
Sintilimab Plus
Anlotinib (n ¼22)
Median age, y (range) 64.5 (4774)
Sex
Male 21 (95.5)
Female 1 (4.5)
Smoking history
Yes 14 (63.6)
No 8 (36.4)
ECOG PS
0 1 (4.5)
1 21 (95.5)
Stage
IIIb 4 (18.2)
IIIc 5 (22.7)
IV 13 (59.1)
BM
Yes 4 (18.2)
No 18 (81.8)
Histology type
Adenocarcinoma 9 (40.9)
Squamous 12 (54.5)
NOS 1 (4.5)
PD-L1 TPS, %
<1 8 (36.4)
149 5 (22.7)
50 8 (36.4)
NE 1 (4.5)
TMB Status
10 Muts/Mb 7 (31.8)
<10 Muts/Mb 11 (50.0)
NE 4 (18.2)
Note. Data are No. (%), unless otherwise noted.
BM, brain metastasis;ECOG PS, Eastern Cooperative Oncology Group per-
formance status; Muts/Mb, mutations per Mb; NE, not evaluable; NOS, not
otherwise specied; PD-L1, programmed death-ligand 1; TMB, tumor mu-
tation burden; TPS, tumor proportion score.
April 2021 Sintilimab Plus Anlotinib in First Line NSCLC 647
treatment-naive patients with advanced NSCLC.
16
Given
that anlotinib targets multiple factors involving tumor
proliferation, vasculature, and tumor microenvironment,
potentially, it could render more benets than VEGF/
VEGF receptor antibodies. More data are being awaited
to conrm this preliminary observation.
Encouraging clinical activity has been observed with
regimens combing ICIs and antiangiogenic tyrosine ki-
nase inhibitors (TKIs) in various advanced solid tumors,
such as pembrolizumab plus axitinib in advanced renal
cell carcinoma,
3
and pembrolizumab plus lenvatinib in
rst-line treatment of advanced liver cancer.
17
A recent
Table 2. Tumor Response
Response
Overall
(n ¼22)
Adenocarcinoma
(n ¼9)
Squamous
(n ¼12)
PD-L1þ
(n ¼13)
PD-L1
(n ¼8)
High TMB
(n ¼7)
Low TMB
(n ¼11)
BOR, n (%)
CR 00 00000
PR 16 (72.7) 8 (88.9) 7 (58.3) 9 (69.2) 6 (75.0) 6 (85.7) 7 (63.6)
Stable disease 6 (27.3) 1 (11.1) 5 (41.7) 4 (30.8) 2 (25.0) 1 (14.3) 4 (36.4)
PD 00 00000
ORR, n (%) 16 (72.7) 8 (88.9) 7 (58.3) 9 (69.2) 6 (75.0) 6 (85.7) 7 (63.6)
95% CI (49.889.3) (51.899.7) (27.784.8) (38.690.9) (34.996.8) (42.199.6) (30.889.1)
DCR, n (%) 22 (100) 9 (100) 12 (100) 13 (100) 8 (100) 7 (100) 11 (100)
95% CI (84.6100) (73.5100) (66.4100) (75.3100) (63.1100) (59.0100) (71.5100)
TTR, mo (95% CI) 1.6 (1.42.9) 1.7 (1.23.2) 1.6 (1.43.1) 1.6 (1.22.9) 1.6 (1.44.2) 1.7 (1.23.2) 1.5 (1.42.4)
DOR, mo (95% CI) NR (3.2NR) NR (2.5NR) NR (3.2NR) NR (3.2NR) NR (2.5NR) NR (NRNR) NR (2.9NR)
PD-L1þwas dened as PD-L1 expression greater than or equal to 1% (Dako 22C3). High TMB was dened as greater than or equal to 10 Mutations/Mb (Foundation
One).
BOR, best overall response; CI, condence interval; CR, complete response; DCR, disease control rate; DOR, duration of response; NR, not reached; ORR,
objective response rate; PD, progressive disease; PD-L1, programmed death-ligand 1; PD-L1þ, PD-L1-positive; PR, partial response; TMB, tumor mutation
burden; TTR, time to response.
Figure 2. PFS. (A) All patients. (B) Nonsquamous versus squamous. (C) By PD-L1 status (positive versus negative). (D) by TMB
status (high versus low). PD-L1positive was dened as PD-L1 expression greater than or equal to 1% (22C3 pharmDx assay).
TMB-H was dened as greater than or equal to 10 Muts/Mb (FoundationOne CDx assay). Adeno, adenocarcinoma; Muts/Mb,
mutations per Mb; PD-L1, programmed death-ligand 1; TMB, tumor mutation burden; TMB-H, TMB-high.
648 Chu et al Journal of Thoracic Oncology Vol. 16 No. 4
phase 1 study reported a 33% ORR in patients with
advanced NSCLC treated with pembrolizumab plus len-
vatinib, in which most patients (18 of 21) enrolled had
received at least one line systemic treatment and 52%
had received previous immunotherapy.
6
This study,
however, enrolled treatment-naive patients only, aiming
to bring this combination strategy in the frontline setting
by reactivating the immunity and remodeling the tumor
microenvironment in the rst place to maximize the
antitumor activity. Our sintilimab plus anlotinib regimen
is the rst combination of an ICI and a multitarget
antiangiogenic TKI as rst-line therapy for patients with
advanced NSCLC, and the efcacy observed was
promising. In addition, because the pembrolizumab/
lenvatinib trial enrolled only non-Asian patients, our
study provided additional evidence for applying this new
strategy in the Chinese population.
In line with the previous reports that immune acti-
vation can lead to durable responses and prolonged
survival in patients with solid tumors,
18-21
a better DOR
and PFS were observed in our study. In a pooled anal-
ysis, the tumor shrinkage rate was suggested to be
positively correlated with PFS and OS in patients with
metastatic NSCLC treated with antiPD-1,
22
and patients
with tumor shrinkage greater than 50% seemed to
benet more. Considering that all patients in this study
have achieved tumor shrinkage and 36.4% (eight of 22)
patients have achieved greater than 50% tumor reduc-
tion, we anticipate that this regimen will render long-
term benets to the patients.
PD-L1 expression level has exhibited a positive cor-
relation with tumor response in previous studies of ICIs
monotherapies.
23,24
Interestingly, in this study, the
response was high regardless of the PD-L1 expression
level, consistent with the observation from the pem-
brolizumab/lenvatinib phase 1b study.
6,23-25
These
ndings suggested that the efcacy of antiPD-1 in
combination with antiangiogenic TKIs could be PD-L1
expressionindependent.
The U.S. Food and Drug Administration recently
approved pembrolizumab for patients with previously
treated unresectable or metastatic TMB-H (10 muta-
tions per Mb) solid tumors who had no satisfactory
alternative treatment options. In our study, TMB-H pa-
tients had an impressive ORR of 85.7%, and no case of
disease progression was reported at the time of data
cutoff, suggesting that the addition of antiangiogenic
TKIs to antiPD-1 immunotherapy may favor the pa-
tients with high genomic instability more. Nevertheless,
the prognostic value of TMB in patients receiving this
regimen should be further veried by larger clinical
studies.
In this study, nine patients (40.9%) developed tumor
cavities, including ve of six patients who achieved sta-
ble disease (Supplementary Figs. 3 and 4). Cavitation is
common in treatment with antiangiogenetic in-
hibitors,
26,27
yet the incidence of the intratumor cavity
with sintilimab and anlotinib is higher than other
antiangiogenic-based therapies (14%24%).
26,27
It is
clear now that both anlotinib and sintilimab can modu-
late the immune-suppressive microenvironment in tu-
mors.
28,29
Thus, sintilimab plus anlotinib may
synergistically enhance the antitumor activity, and is
more likely to lead to the development of tumor cavi-
tation. A study of anlotinib in NSCLC indicated that pa-
tients with tumor cavities had a better prognosis than
those without cavities.
30
In our study, nine patients
Table 3. TRAEs
AEs
Any Grade
No. (%)
Grade 3
No. (%)
Hemorrhage 13 (59.1) 0 (0.0)
Urinary occult blood 10 (45.5) 0 (0.0)
Hemoptysis 3 (13.6) 0 (0.0)
Fecal occult blood 2 (9.1) 0 (0.0)
Epistaxis 1 (4.5) 0 (0.0)
Gingival bleeding 1 (4.5) 0 (0.0)
Hypothyroidism 11 (50.0) 0 (0.0)
Uric acid increased 9 (40.9) 0 (0.0)
Hand-foot skin reaction 8 (36.4) 1 (4.5)
Hypoalbuminemia 8 (36.4) 0 (0.0)
Hypertension 7 (31.8) 2 (9.1)
ALT increased 7 (31.8) 0 (0.0)
Direct bilirubin increased 7 (31.8) 0 (0.0)
Rash 5 (22.7) 1 (4.5)
Pneumonitis 5 (22.7) 0 (0.0)
Immune-related pneumonitis
a
3 (13.6) 0 (0.0)
Fever 5 (22.7) 0 (0.0)
Total bilirubin increased 5 (22.7) 0 (0.0)
Hyponatremia 4 (18.2) 1 (4.5)
Proteinuria 4 (18.2) 1 (4.5)
Diarrhea 4 (18.2) 1 (4.5)
Hypochloremia 4 (18.2) 0 (0.0)
Fatigue 4 (18.2) 0 (0.0)
Oral ulcer 4 (18.2) 0 (0.0)
AST increased 4 (18.2) 0 (0.0)
Hoarseness 4 (18.2) 0 (0.0)
Anorexia 4 (18.2) 0 (0.0)
Gum pain 4 (18.2) 0 (0.0)
Lipase elevation 3 (18.2) 1 (4.5)
Amylase increased 2 (9.1) 1 (4.5)
Immune-related myositis
b
1 (4.5) 1 (4.5)
Cerebral infarction 1 (4.5) 1 (4.5)
Blood cortisol decreased 1 (4.5) 1 (4.5)
Adrenal insufciency
b
1 (4.5) 1 (4.5)
TRAEs occurring at any grade in more than 15% of patients or grade 3 or worse
in more than 1% of patients in the safety population are illustrated in this
table.
a
One grade 5 event was observed (immune-related pneumonitis).
b
Immune-related myositis and adrenal insufciency occurred in the same
patient. No grade 4 events were observed.
AE, adverse event; AST, aspartate aminotransferase; ALT, alanine amino-
transferase; TRAE, treatment-related AE.
April 2021 Sintilimab Plus Anlotinib in First Line NSCLC 649
developed tumor cavities, and they tended to have du-
rable responses (Supplementary Fig. 4). Of note, only
one experienced grade 1 hemoptysis among these pa-
tients, and no other pulmonary hemorrhage events were
observed.
Unlike the IMpower150 phase 3 study,
8
which only
enrolled patients with nonsquamous NSCLC, 54.5% of
patients in this study had squamous cell carcinomas.
Therefore, a signicant clinical advantage noted with
sintilimab plus anlotinib is that it can benet both his-
tologic subgroups. This is consistent with the previous
conclusions that the benets of anlotinib monotherapy
or sintilimab-based chemotherapy are regardless of
NSCLC histology type.
9,14
Among the four patients with BM at baseline, three
achieved overall PR, and one achieved stable disease.
The observed intracranial and systemic responses were
comparable, indicating that the regimen of sintilimab
plus anlotinib had synergistic effects in the brain. This
nding was consistent with the phase 2 trial of patients
with NSCLC with BM
31
and the KEYNOTE-189 posthoc
analysis.
32
Safety was found to be consistent with known safety
proles of either sintilimab or anlotinib alone.
12,14
No
new safety signal was identied.
10,11,33
Grade 3 or higher
TRAEs occurred in 54.5% of patients, whereas the inci-
dence of grade 3 or higher TRAEs was generally above
60% in chemotherapy-containing immunother-
apies.
8,34,35
Except for two cases of grade 3 hyperten-
sion, most grade 3 TRAEs occurred only once. Five
patients experienced grade greater than or equal to 3
TRAEs 1 year after the treatment, which might be
associated with the long treatment duration. Although
hypothyroidism occurred in half of the patients in this
study, most subjects had grade 1 to 2 and recovered
without medication. A similar hypothyroidism incidence
was observed in patients treated with pembrolizumab
plus lenvatinib (47%) or axitinib (35.4%).
3,36
The major limitation of this hypothesis-generating,
single-center study is the relatively small sample size.
To further evaluate this hypothesis, a multicenter, ran-
domized, phase 2 trial (NCT04124731) is underway to
generate more clinical evidence on efcacy, safety,
quality of life, and translational biomarkers. We also
noted that some of the baseline characteristics in this
study were not representative of the prevalence of pa-
tients with advanced NSCLC in the Peoples Republic of
China, with men and squamous cell carcinoma histology
type predominating in the enrolled patients. The most
likely reason for this was the exclusion of EGFR muta-
tions, which were more common in Asian women and
adenocarcinoma types than in men and squamous types.
In summary, the combination of sintilimab plus
anlotinib has exhibited encouraging efcacy, durability,
and tolerability in this phase 1b clinical trial, making it a
promising chemotherapy-free option for rst-line treat-
ment in patients with advanced NSCLC. This novel
combination has potential efcacy for a broader range of
patients with NSCLC, regardless of histologic subtype or
PD-L1 status. In addition, its favorable and manageable
safety prole may provide an improved quality of life
and offer potential opportunities for subsequent treat-
ments. These preliminary ndings have exhibited the
worthiness of the development of sintilimab plus anlo-
tinib in rst-line advanced NSCLC.
Acknowledgments
This study was supported by the Western Medicine
Guidance Project of Shanghai Science and Technology
Commission (No. 18411968500), the Multicenter Clin-
ical Research Project of Shanghai Jiao Tong University
Medical School (No. DLY201816), and the Cooperative
Innovation Project of Shanghai Chest Hospital (No.
YJXT20190102). The study drugs of sintilimab and
anlotinib were provided by Innovent (Suzhou) Biophar-
maceutical Co., Ltd. and Chia Tai Tianqing Pharmaceu-
tical Group Co., Ltd., respectively. The authors would like
to thank all patients who participated in the study and
the clinical personnel involved in data collection. The
funders have no role in study design; in the collection,
analysis, and interpretation of data; in the writing of the
report; and in the decision to submit the article for
publication.
References
1. Khan KA, Kerbel RS. Improving immunotherapy outcomes
with anti-angiogenic treatments and vice versa. Nat Rev
Clin Oncol. 2018;15:310324.
2. Tian L, Goldstein A, Wang H, et al. Mutual regulation of
tumour vessel normalization and immunostimulatory
reprogramming. Nature. 2017;544:250254.
3. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus
axitinib versus sunitinib for advanced renal-cell carci-
noma. N Engl J Med. 2019;380:11161127.
4. McDermott DF, Huseni MA, Atkins MB, et al. Clinical ac-
tivity and molecular correlates of response to atezoli-
zumab alone or in combination with bevacizumab versus
sunitinib in renal cell carcinoma. Nat Med. 2018;24:749
757.
5. Herbst RS, Arkenau HT, Santana-Davila R, et al. Ramu-
cirumab 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;20:11091123.
6. Taylor MH, Lee CH, Makker V, et al. Phase IB/II trial of
lenvatinib plus pembrolizumab in patients with
advanced renal cell carcinoma, endometrial cancer, and
other selected advanced solid tumors. J Clin Oncol.
2020;38:11541163.
650 Chu et al Journal of Thoracic Oncology Vol. 16 No. 4
7. Makker V, Taylor MH, Aghajanian C, et al. Lenvatinib plus
pembrolizumab in patients with advanced endometrial
cancer. J Clin Oncol. 2020;38:29812992.
8. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab
for rst-line treatment of metastatic nonsquamous
NSCLC. N Engl J Med. 2018;378:22882301.
9. Xu N, Ying K, Wang Z, et al. Phase Ib study of sintilimab
in combination with chemotherapy for 1L advanced or
metastatic non-small cell lung cancer (NSCLC). J Clin
Oncol. 2019;37(suppl 15):e20546e20546.
10. Hoy SM. Sintilimab: rst global approval. Drugs.
2019;79:341346.
11. Yang Y, Wang Z, Fang J, et al. Efcacy and Safety of
Sintilimab Plus pemetrexed and platinum as rst-line
treatment for locally advanced or metastatic non-
squamous NSCLC: a randomized, double-blind, phase 3
study (Oncology pRogram by InnovENT anti-PD-1-11).
J Thorac Oncol. 2020;15:16361646.
12. Gao S, Li N, Gao S, et al. Neoadjuvant PD-1 inhibitor
(Sintilimab) in NSCLC. J Thorac Oncol. 2020;15:816826.
13. Shen G, Zheng F, Ren D, et al. Anlotinib: a novel multi-
targeting tyrosine kinase inhibitor in clinical develop-
ment. J Hematol Oncol. 2018;11:120.
14. 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 [published
correction appears in JAMA Oncol. 2018;4:1625]. JAMA
Oncol. 2018;4:15691575.
15. 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;7:387401.
16. Herbst R, Arkenau H, Bendell J, et al. MA14.07 phase I
expansion cohort of ramucirumab plus pembrolizumab in
advanced treatment-naïve non-small cell lung cancer
(JVDF). J Thorac Oncol. 2019;14:S307.
17. Llovet J, Shepard KV, Finn RS, et al. A phase Ib trial of
lenvatinib (LEN) plus pembrolizumab (PEMBRO) in unre-
sectable hepatocellular carcinoma (uHCC): updated re-
sults. Ann Oncol. 2019;30(suppl 5):v286v287.
18. Cremolini C, Loupakis F, Antoniotti C, et al. Early tumor
shrinkage and depth of response predict long-term
outcome in metastatic colorectal cancer patients
treated with rst-line chemotherapy plus bevacizumab:
results from phase III TRIBE trial by the Gruppo Oncolo-
gico del Nord Ovest. Ann Oncol. 2015;26:11881194.
19. Grünwald V, Lin X, Kalanovic D, Simantov R. Early tumour
shrinkage: a tool for the detection of early clinical ac-
tivity in metastatic renal cell carcinoma. Eur Urol.
2016;70:10061015.
20. Nishina T, Azuma M, Nishikawa K, et al. Early tumor
shrinkage and depth of response in patients with
advanced gastric cancer: a retrospective analysis of a
randomized phase III study of rst-line S-1 plus oxali-
platin vs. S-1 plus cisplatin. Gastric Cancer. 2019;22:138
146.
21. Vivaldi C, Fornaro L, Cappelli C, et al. Early tumor
shrinkage and depth of response evaluation in metasta-
tic pancreatic cancer treated with rst line chemo-
therapy: an observational retrospective cohort study.
Cancers (Basel). 2019;11:939.
22. McCoach CE, Blumenthal GM, Zhang L, et al. Exploratory
analysis of the association of depth of response and
survival in patients with metastatic non-small-cell lung
cancer treated with a targeted therapy or immuno-
therapy. Ann Oncol. 2017;28:27072714.
23. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab
versus chemotherapy for previously untreated, PD-
L1-expressing, locally advanced or metastatic non-
small-cell lung cancer (KEYNOTE-042): a randomised,
open-label, controlled, phase 3 trial. Lancet. 2019;
393:18191830.
24. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pem-
brolizumab versus chemotherapy for PD-L1-positive
non-small-cell lung cancer. N Engl J Med. 2016;
375:18231833.
25. Reck M, Rodríguez-Abreu D, Robinson AG, et al.
Updated analysis of KEYNOTE-024: pembrolizumab
versus platinum-based chemotherapy for advanced
non-small-cell lung cancer with PD-L1 tumor propor-
tion score of 50% or greater. JClinOncol.
2019;37:537546.
26. Jiang M, Zhang C, Liu D, et al. Inuence and mechanism
of lung cavitation development on antiangiogenic ther-
apy. Transl Lung Cancer Res. 2019;8:500512.
27. Crabb SJ, Patsios D, Sauerbrei E, et al. Tumor cavitation:
impact on objective response evaluation in trials of
angiogenesis inhibitors in non-small-cell lung cancer.
J Clin Oncol. 2009;27:404410.
28. Liu S, Qin T, Liu Z, et al. Anlotinib alters tumor immune
microenvironment by downregulating PD-L1 expression
on vascular endothelial cells. Cell Death Dis.
2020;11:309.
29. Wang J, Fei K, Jing H, et al. Durable blockade of PD-1
signaling links preclinical efcacy of sintilimab to its
clinical benet. MAbs. 2019;11:14431451.
30. Chen D, Xu J, Zhao Y, et al. Prognostic value of tumor
cavitation in extensive-stage small-cell lung cancer
patients treated with anlotinib. J Cancer Res Clin Oncol.
2020;146:401406.
31. Goldberg SB, Schalper KA, Gettinger SN, et al. Pem-
brolizumab for management of patients with NSCLC and
brain metastases: long-term results and biomarker
analysis from a non-randomised, open-label, phase 2
trial. Lancet Oncol. 2020;21:655663.
32. Gadgeel S, Rodriguez-Abreu D, Speranza G, et al.
Updated analysis from KEYNOTE-189: pembrolizumab or
placebo plus pemetrexed and platinum for previously
untreated metastatic nonsquamous non-small-cell lung
cancer. J Clin Oncol. 2020;38:15051517.
33. Han B, Li K, Zhao Y, et al. Anlotinib as a third-line
therapy in patients with refractory advanced non-
small-cell lung cancer: a multicentre, randomised
phase II trial (ALTER0302). Br J Cancer. 2018;118:654
661.
April 2021 Sintilimab Plus Anlotinib in First Line NSCLC 651
34. Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pem-
brolizumab plus chemotherapy in metastatic non-
small-cell lung cancer. N Engl J Med. 2018;378:
20782092.
35. West H, McCleod M, Hussein M, et al. Atezolizumab in
combination with carboplatin plus nab-paclitaxel
chemotherapy compared with chemotherapy alone as
rst-line treatment for metastatic non-squamous
non-small-cell lung cancer (IMpower130): a multicentre,
randomised, open-label, phase 3 trial. Lancet Oncol.
2019;20:924937.
36. Makker V, Rasco D, Vogelzang NJ, et al. Lenvatinib plus
pembrolizumab in patients with advanced endometrial
cancer: an interim analysis of a multicentre, open-label,
single-arm, phase 2 trial. Lancet Oncol. 2019;20:711
718.
652 Chu et al Journal of Thoracic Oncology Vol. 16 No. 4
... Preclinical studies have shown the potential synergistic effect of this combination. [18][19][20] Antiangiogenic drugs can not only reverse the immunosuppressive effect caused by vascular endothelial growth factor (VEGF), but also normalize the tumor vascular system and promote the delivery of T cells and other immune effector molecules. [21][22][23] Moreover, ICIs can normalize the tumor vascular system and increase the infiltration and killing function of effector T cells by activating them. ...
... ICI in combination with anlotinib was well-tolerated and showed clinical efficacy as first-line treatment for patients with stage IV NSCLC. 19,25 The phase 2 WJOG10718L study showed a promising objective response rate (ORR) and disease control rate (DCR) with first-line atezolizumab plus bevacizumab in patients with non-squamous NSCLC. 26 Nevertheless, the efficacy and safety of this combination strategy as later-line therapy in patients with ECOG PS 2 remain unclear. ...
Article
Full-text available
Background Patients with Eastern Cooperative Oncology Group performance status (ECOG PS) 2 probably cannot tolerate chemotherapy or other antitumor therapies. Some studies have reported that immunotherapy combined with antiangiogenic therapy is well‐tolerated and shows good antitumor activity. However, the efficacy of this combination as a later‐line therapy in patients with ECOG PS 2 is unclear. This study evaluated the effectiveness and safety of this combination strategy as third‐ or further‐line therapy in stage IV non‐small cell lung cancer (NSCLC) patients with ECOG PS 2. Methods In this retrospective study, patients treated with camrelizumab plus antiangiogenic therapy (bevacizumab, anlotinib, or recombinant human endostatin) were included. Objective response rate (ORR), disease control rate (DCR), progression‐free survival (PFS), overall survival (OS), quality of life (QOL) assessed by ECOG PS, and safety were analyzed. Results Between January 10, 2019, and February 28, 2024, a total of 59 patients were included. The ORR was 35.6% (21/59) and the DCR was 86.4%. With a median follow‐up of 10.5 months (range: 0.7–23.7), the median PFS was 5.5 months (95% confidence interval [CI]: 3.8–7.3) and the median OS was 10.5 months (95% CI: 11.2–13.6). QOL was improved (≥1 reduction in ECOG PS) in 39 patients (66.1%). The most common Grade 3–4 treatment‐related adverse events were hepatic dysfunction (6 [10%]), hypertension (5 [8%]), and hypothyroidism (3 [5%]). There were no treatment‐related deaths. Conclusions Third‐ or further‐line immunotherapy combined with antiangiogenic therapy is well‐tolerated and shows good antitumor activity in stage IV NSCLC patients with ECOG PS 2. Future large‐scale prospective studies are required to confirm the clinical benefits of this combination therapy.
... The targets of anlotinib include VEGFR, fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor (PDGFR) and cKit [13]. In our previous phase Ib study, the combination of anlotinib with sintilimab, a fully humanized PD-1 monoclonal antibody, showed a potent synergetic effect in treatment-naïve advanced NSCLC patients without driver gene alterations [14]. The combination conferred an objective response rate (ORR) of 72.7% and a median progression-free survival (PFS) of 15 months [14]. ...
... In our previous phase Ib study, the combination of anlotinib with sintilimab, a fully humanized PD-1 monoclonal antibody, showed a potent synergetic effect in treatment-naïve advanced NSCLC patients without driver gene alterations [14]. The combination conferred an objective response rate (ORR) of 72.7% and a median progression-free survival (PFS) of 15 months [14]. ...
Article
Full-text available
Background Resistance to immune checkpoint inhibitors (ICIs) represents a major unmet medical need in non-small cell lung cancer (NSCLC) patients. Vascular endothelial growth factor (VEGF) inhibition may reverse a suppressive microenvironment and recover sensitivity to subsequent ICIs. Methods This phase Ib/IIa, single-arm study, comprised dose-finding (Part A) and expansion (Part B) cohorts. Patients with ICIs-refractory NSCLC were enrolled to receive anlotinib (a multi-target tyrosine kinase inhibitor) orally (from days 1 to 14 in a 21-day cycle) and nivolumab (360 mg every 3 weeks, intravenously) on a 21-day treatment cycle. The first 21-day treatment cycle was a safety observation period (phase Ib) followed by a phase II expansion cohort. The primary objectives were recommended phase 2 dose (RP2D, part A), safety (part B), and objective response rate (ORR, part B), respectively. Results Between November 2020 and March 2022, 34 patients were screened, and 21 eligible patients were enrolled (6 patients in Part A). The RP2D of anlotinib is 12 mg/day orally (14 days on and 7 days off) and nivolumab (360 mg every 3 weeks). Adverse events (AEs) of any cause and treatment-related AEs (TRAEs) were reported in all treated patients. Two patients (9.5%) experienced grade 3 TRAE. No grade 4 or higher AEs were observed. Serious AEs were reported in 4 patients. Six patients experienced anlotinib interruption and 4 patients experienced nivolumab interruption due to TRAEs. ORR and disease control rate (DCR) was 19.0% and 76.2%, respectively. Median PFS and OS were 7.4 months (95% CI, 4.3-NE) and 15.2 months (95% CI, 12.1-NE), respectively. Conclusion Our study suggests that anlotinib combined with nivolumab shows manageable safety and promising efficacy signals. Further studies are warranted. Trial registration NCT04507906 August 11, 2020.
... Recombinant human endostatin, an antagonist of vascular endothelial growth factor, inhibits tumor angiogenesis through multiple pathways and has shown promising results in treating various solid tumors. Its potential as a combination therapy with immunotherapy is appealing [8]. A recent study compared the clinical efficacy of two immune combination therapies: immunothera-py combined with recombinant human endostatin and immunotherapy combined with chemotherapy, as second-line treatments for advanced NSCLC [9]. ...
... Some studies have shown that the combination of anlotinib and ICIs has a synergistic effect. For patients with advanced NSCLC without epidermal growth factor receptor (EGFR) mutations, the combination of anlotinib and ICIs has good e cacy and tolerance as rst-line therapy (Chu et al. 2021). In second-line and beyond, anlotinib plus ICIs produced higher objective effectiveness rates (ORR) and longer progression-free survival (PFS) compared to ICIs monotherapy ). ...
Preprint
Full-text available
Purpose: The combination of anlotinib with immune checkpoint inhibitors (ICIs) has become a common treatment modality in clinical practice. However, the optimal dose of anlotinib to use remains unclear. Methods: We collected patients with advanced non-small cell lung cancer (NSCLC) who received programmed cell death-1 (PD-1) or programmed cell death ligand 1 (PD-L1) blockade combined with different dose anlotinib as second-line or later line therapy. Subsequently, the efficacy and safety of the combination therapy as well as subgroup analyses of different doses of anlotinib were analyzed. Cox regression was performed to analyze significant factors correlated with progression-free survival (PFS) and overall survival (OS). Results: A total of 50 eligible patients with NSCLC who received anlotinib combined with ICIs therapy were included, of which 27 received low-dose anlotinib (8 mg), and 23 were administered high-dose anlotinib (12 mg). The median PFS (mPFS) and the median OS (mOS) for all patients were 8.3 months (95% CI 6.3–10.3) and 17.6 months (95% CI 16.5–18.7), respectively. Subgroup analyses showed that patients treated with 8 mg of anlotinib plus ICIs had significantly longer mPFS than those treated with 12 mg of amlotinib plus ICIs (8.7 months vs 6.7 months; p=0.016). The overall incidence of adverse events (AEs) was 68.0%, and the most common AEs of all grades were hypertension. Meanwhile, the incidence of adverse events was higher for 12 mg of anlotinib plus ICIs than that of 8 mg of anlotinib plus ICIs(82.6% vs 55.6%, P = 0.041). Conclusion: Low-dose anlotinib in combination with ICIs for advanced NSCLC may be an effective and well-tolerated option.
... Clinically, anlotinib plus ICIs shows higher response than ICIs monotherapy in NSCLC, but with more adverse events (19). A phase II study reported efficacy, durability, and safety for sintilimab plus anlotinib (20), while a real-world study found no PFS difference between anlotinib plus ICIs versus anlotinib monotherapy (21). Collectively, current evidence suggests anlotinib plus ICIs may be more effective than monotherapy in NSCLC. ...
Article
Full-text available
Background Non-small cell lung cancer (NSCLC) remains a leading cause of cancer mortality. Combined anlotinib and immune checkpoint inhibitors (ICIs) therapy may have synergistic antitumor effects in NSCLC. This study aimed to comparing the efficacy and safety of anlotinib and ICIs treatment, monotherapy and combination in NSCLC. Methods We performed a systematic review and network meta-analysis of 14 studies involving 4,308 NSCLC patients across four regimens: anlotinib, ICIs, anlotinib plus ICIs, and placebo. Efficacy outcomes were progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR). Safety outcomes included treatment-related adverse events (TRAEs), TRAE grade three or higher (TRAE ≥3). Analyses were performed in RevMan 5.3 and R 3.5.1 (gemtc package). P<0.05 or effect estimate with 95% confidence interval (CI) that did not include 1 indicated statistical significance. Results Fourteen publications involving 4,308 patients across four treatment regimens (anlotinib, ICIs, anlotinib plus ICIs, placebo) were included. For PFS, network meta-analysis showed all three interventions significantly improved PFS versus placebo. Anlotinib plus ICIs demonstrated the greatest PFS improvement [hazard ratio (HR) =0.24; 95% CI: 0.14, 0.36], followed by anlotinib (HR =0.37; 95% CI: 0.23, 0.58), and ICIs (HR =0.43; 95% CI: 0.27, 0.67). For OS, compared to placebo, anlotinib plus ICIs showed the greatest OS improvement (HR =0.52; 95% CI: 0.33, 0.74), followed by anlotinib (HR =0.66; 95% CI: 0.47, 0.95), and ICIs (HR =0.72; 95% CI: 0.54, 0.97). For ORR, anlotinib plus ICIs demonstrated the greatest improvement versus placebo [odds ratio (OR) =5.29; 95% CI: 3.32, 8.58], followed by anlotinib (OR =4.38; 95% CI: 2.42, 8.19), and ICIs (OR =2.17; 95% CI: 1.65, 2.89). For DCR, anlotinib plus ICIs showed the greatest improvement versus placebo (OR =13.32; 95% CI: 4.99, 45.09), followed by anlotinib (OR =5.56; 95% CI: 2.17, 14.38), and ICIs (OR =3.46; 95% CI: 1.29, 10.85). Compared to placebo, anlotinib was associated with the highest risk of TRAEs (OR =3.67, 95% CI: 1.12, 15.77), followed by ICIs (OR =1.83; 95% CI: 1.26, 2.69). Due to lack of data on anlotinib plus ICIs, no comparison was conducted. For grade ≥3 TRAEs, compared to placebo, anlotinib increased the risk (OR =3.67; 95% CI: 1.12, 15.77), while anlotinib plus ICIs (OR =2.45; 95% CI: 0.51, 11.6) and ICIs (OR =1.29; 95% CI: 0.33, 4.38) did not increase the risk. Conclusions Anlotinib combined with ICIs demonstrates improved efficacy over monotherapy for NSCLC treatment, without increased adverse events.
... Among them, ICIs combined with anti-angiogenic drugs have achieved gratifying results in the treatment of NSCLC, showing good efficacy and acceptable tolerance, and have certain clinical application prospects. It has become a hotspot in the research of anti-tumor programs (6)(7)(8)(9)(10). ...
... Moreover, anlotinib improves the immune microenvironment by downregulating PD-L1 expression in BECs in vivo 11 . A clinical study using the combination of anlotinib and the programmed death PD-1 antibody, sintilimab, as the first-line treatment for advanced NSCLC revealed good efficacy 16 . We hypothesized that the mechanisms underlying the reinforcement of ICIs may be attributed to the downregulation of PD-L1 in the microconduit endothelium inside the tumor (Figure 1). ...
Article
Objective: The possible enhancing effect of anlotinib on programmed death receptor ligand (PD-L1) antibody and the efficacy-predicting power of PD-L1 in micro-conduit endothelium, including lymphatic endothelial cells (LECs) and blood endothelial cells (BECs), were determined to identify patients who would benefit from this treatment. Methods: PD-L1 positivity in LECs, BECs, and tumor cells (TCs) was assessed using paraffin sections with multicolor immunofluorescence in an investigator’s brochure clinical trial of TQB2450 (PD-L1 antibody) alone or in combination with anlotinib in patients with non-small cell lung cancer. Progression-free survival (PFS) with different levels of PD-L1 expression was compared between the two groups. Results: Among 75 patients, the median PFS (mPFS) was longer in patients who received TQB2450 with anlotinib [10 and 12 mg (161 and 194 days, respectively)] than patients receiving TQB2450 alone (61 days) [hazard ratio (HR)10 mg = 0.390 (95% confidence interval {CI}, 0.201–0.756), P = 0.005; HR12 mg = 0.397 (0.208–0.756), P = 0.005]. The results were similar among 58 patients with high PD-L1 expression in LECs and TCs [159 and 209 vs. 82 days, HR10 mg = 0.445 (0.210–0.939), P = 0.034; HR12 mg = 0.369 (0.174–0.784), P = 0.009], and 53 patients with high PD-L1 expression in BECs and TCs [161 and 209 vs. 41 days, HR10 mg = 0.340 (0.156–0.742), P = 0.007; HR12 mg = 0.340 (0.159–0.727), P = 0.005]. No differences were detected in the mPFS between the TQB2450 and combination therapy groups in 13 low/no LEC-expressing and 18 low/no BEC-expressing PD-L1 cases. Conclusions: Mono-immunotherapy is not effective in patients with high PD-L1 expression in LECs and/or BECs. Anlotinib may increase efficacy by downregulating PD-L1 expression in LECs and/or BECs, which is presumed to be a feasible marker for screening the optimal immune patient population undergoing anti-angiogenic therapy.
Article
Full-text available
Nowadays, immunotherapy is one of the most promising anti-tumor therapeutic strategy. Specifically, immune-related targets can be used to predict the efficacy and side effects of immunotherapy and monitor the tumor immune response. In the past few decades, increasing numbers of novel immune biomarkers have been found to participate in certain links of the tumor immunity to contribute to the formation of immunosuppression and have entered clinical trials. Here, we systematically reviewed the oncogenesis and progression of cancer in the view of anti-tumor immunity, particularly in terms of tumor antigen expression (related to tumor immunogenicity) and tumor innate immunity to complement the cancer-immune cycle. From the perspective of integrated management of chronic cancer, we also appraised emerging factors affecting tumor immunity (including metabolic, microbial, and exercise-related markers). We finally summarized the clinical studies and applications based on immune biomarkers. Overall, immune biomarkers participate in promoting the development of more precise and individualized immunotherapy by predicting, monitoring, and regulating tumor immune response. Therefore, targeting immune biomarkers may lead to the development of innovative clinical applications.
Article
Full-text available
Introduction Sintilimab, an anti-programmed death 1 antibody, plus pemetrexed and platinum had shown promising efficacy for nonsquamous non-small cell lung cancer in a phase 1b study. We conducted a randomized, double-blind, phase 3 study to compare the efficacy and safety of sintilimab with placebo, both in combination with such chemotherapy. (ClinicalTrials.gov: NCT03607539) Methods A total of 397 patients with previously untreated locally advanced or metastatic nonsquamous NSCLC without sensitizing epidermal growth factor receptor or anaplastic lymphoma kinase genomic aberration were randomized (2:1 ratio) to receive either sintilimab 200 mg or placebo plus pemetrexed and platinum Q3W for 4 cycles, followed by sintilimab or placebo plus pemetrexed therapy. Crossover or treatment beyond disease progression was allowed. The primary endpoint was progression-free survival (PFS) by independent radiographic review committee. Results As of Nov. 15, 2019, the median follow-up was 8.9 months. The median PFS was significantly longer in the sintilimab-combination group than that in the placebo-combination group (8.9 vs. 5.0 months; HR, 0.482, 95%CI, 0.362 to 0.643; p < 0.00001). The confirmed objective response rate was 51.9 % (95% CI, 45.7% to 58.0%) in sintilimab-combination group and 29.8% (95% CI, 22.1% to 38.4%) in placebo-combination group. The incidence of grade 3 or higher adverse events was 61.7% in sintilimab-combination group and 58.8% in placebo-combination group. Conclusions In Chinese patients with previously untreated locally advanced or metastatic nonsquamous NSCLC, the addition of sintilimab to chemotherapy of pemetrexed and platinum resulted in significantly longer PFS than that of chemotherapy alone with manageable safety profiles.
Article
Full-text available
Aberrant vascular network is a hallmark of cancer. However, the role of vascular endothelial cells (VECs)-expressing PD-L1 in tumor immune microenvironment and antiangiogenic therapy remains unclear. In this study, we used the specimens of cancer patients for immunohistochemical staining to observe the number of PD-L1+ CD34+ VECs and infiltrated immune cells inside tumor specimens. Immunofluorescence staining and flow cytometry were performed to observe the infiltration of CD8+ T cells and FoxP3+ T cells in tumor tissues. Here, we found that PD-L1 expression on VECs determined CD8+ T cells’, FoxP3+ T cells’ infiltration, and the prognosis of patients with lung adenocarcinoma. Anlotinib downregulated PD-L1 expression on VECs through the inactivation of AKT pathway, thereby improving the ratio of CD8/FoxP3 inside tumor and remolding the immune microenvironment. In conclusion, our results demonstrate that PD-L1 high expression on VECs inhibits the infiltration of CD8+ T cells, whereas promotes the aggregation of FoxP3+ T cells into tumor tissues, thus becoming an “immunosuppressive barrier”. Anlotinib can ameliorate the immuno-microenvironment by downregulating PD-L1 expression on VECs to inhibit tumor growth.
Article
Full-text available
PURPOSE Patients with advanced endometrial carcinoma have limited treatment options. We report final primary efficacy analysis results for a patient cohort with advanced endometrial carcinoma receiving lenvatinib plus pembrolizumab in an ongoing phase Ib/II study of selected solid tumors. METHODS Patients took lenvatinib 20 mg once daily orally plus pembrolizumab 200 mg intravenously once every 3 weeks, in 3-week cycles. The primary end point was objective response rate (ORR) at 24 weeks (ORR Wk24 ); secondary efficacy end points included duration of response (DOR), progression-free survival (PFS), and overall survival (OS). Tumor assessments were evaluated by investigators per immune-related RECIST. RESULTS At data cutoff, 108 patients with previously treated endometrial carcinoma were enrolled, with a median follow-up of 18.7 months. The ORR Wk24 was 38.0% (95% CI, 28.8% to 47.8%). Among subgroups, the ORR Wk24 (95% CI) was 63.6% (30.8% to 89.1%) in patients with microsatellite instability (MSI)–high tumors (n = 11) and 36.2% (26.5% to 46.7%) in patients with microsatellite-stable tumors (n = 94). For previously treated patients, regardless of tumor MSI status, the median DOR was 21.2 months (95% CI, 7.6 months to not estimable), median PFS was 7.4 months (95% CI, 5.3 to 8.7 months), and median OS was 16.7 months (15.0 months to not estimable). Grade 3 or 4 treatment-related adverse events occurred in 83/124 (66.9%) patients. CONCLUSION Lenvatinib plus pembrolizumab showed promising antitumor activity in patients with advanced endometrial carcinoma who have experienced disease progression after prior systemic therapy, regardless of tumor MSI status. The combination therapy had a manageable toxicity profile.
Article
Full-text available
PURPOSE In KEYNOTE-189, first-line pembrolizumab plus pemetrexed-platinum significantly improved overall survival (OS) and progression-free survival (PFS) compared with placebo plus pemetrexed-platinum in patients with metastatic nonsquamous non‒small-cell lung cancer (NSCLC), irrespective of tumor programmed death-ligand 1 (PD-L1) expression. We report an updated analysis from KEYNOTE-189 (ClinicalTrials.gov: NCT02578680 ). METHODS Patients were randomly assigned (2:1) to receive pemetrexed and platinum plus pembrolizumab (n = 410) or placebo (n = 206) every 3 weeks for 4 cycles, then pemetrexed maintenance plus pembrolizumab or placebo for up to a total of 35 cycles. Eligible patients with disease progression in the placebo-combination group could cross over to pembrolizumab monotherapy. Response was assessed per RECIST (version 1.1) by central review. No alpha was assigned to this updated analysis. RESULTS As of September 21, 2018 (median follow-up, 23.1 months), the updated median (95% CI) OS was 22.0 (19.5 to 25.2) months in the pembrolizumab-combination group versus 10.7 (8.7 to 13.6) months in the placebo-combination group (hazard ratio [HR], 0.56; 95% CI, 0.45 to 0.70]). Median (95% CI) PFS was 9.0 (8.1 to 9.9) months and 4.9 (4.7 to 5.5) months, respectively (HR, 0.48; 95% CI, 0.40 to 0.58). Median (95% CI) time from randomization to objective tumor progression on next-line treatment or death from any cause, whichever occurred first (progression-free-survival-2; PFS-2) was 17.0 (15.1 to 19.4) months and 9.0 (7.6 to 10.4) months, respectively (HR, 0.49; 95% CI, 0.40 to 0.59). OS and PFS benefits with pembrolizumab were observed regardless of PD-L1 expression or presence of liver/brain metastases. Incidence of grade 3-5 adverse events was similar in the pembrolizumab-combination (71.9%) and placebo-combination (66.8%) groups. CONCLUSION First-line pembrolizumab plus pemetrexed-platinum continued to demonstrate substantially improved OS and PFS in metastatic nonsquamous NSCLC, regardless of PD-L1 expression or liver/brain metastases, with manageable safety and tolerability.
Article
Full-text available
Background: Programmed death receptor-1 (PD-1) inhibitors have shown efficacy in first line treatment of non-small cell lung cancer (NSCLC), however, evidence of PD-1 inhibitor as neoadjuvant treatment is limited. This is a phase 1b study to evaluate the safety and outcome of PD-1 inhibitor in neoadjuvant setting. Methods: Treatment-naïve patients with resectable NSCLC (stage IA-IIIB) received two cycles of sintilimab (200 mg, intravenously, day 1/22). Surgery was performed between day 29-43. PET-CT was obtained at baseline and prior to surgery. Primary endpoint was safety. Efficacy endpoints included rate of major pathologic response (MPR) and objective response rate (ORR). PD-L1 expression was also evaluated. (Registration Number: ChiCTR-OIC-17013726). Results: Forty patients enrolled, all of whom received 2 doses of sintilimab, and 37 underwent radical resection. Twenty-one (52.5%) patients experienced neoadjuvant treatment-related adverse events (TRAEs). Four (10.0%) patients experienced grade 3-4 neoadjuvant TRAEs, and one patient had grade 5 TRAE. Eight patients achieved radiological partial response, resulting in an ORR of 20.0%. Among 37 patients, 15 (40.5%) achieved MPR, including 6 (16.2%) with a pathologic complete response in primary tumor and 3 (8.1%) in lymph nodes as well. Squamouse cell NSCLC exhibited superior response compared to adenocarcinoma (MPR: 48.4% VS. 0%). Decrease of maximum standardized uptake values (SUVmax) after sintilimab treatment correlated with pathological remission (p<0.00001). Baseline PD-L1 expression of stromal cells instead of tumor cells was correlated with pathological regression (p=0.0471). Conclusion: Neoadjuvant sintilimab was tolerable for NSCLC patients and 40.5% MPR rate is encouraging. The decrease of SUVmax after sintilimab might predict pathologic response.
Article
Full-text available
PURPOSE Modulation of vascular endothelial growth factor–mediated immune suppression via angiogenesis inhibition may augment the activity of immune checkpoint inhibitors. We report results from the dose-finding and initial phase II expansion of a phase Ib/II study of lenvatinib plus pembrolizumab in patients with selected advanced solid tumors. METHODS Eligible patients had metastatic renal cell carcinoma (RCC), endometrial cancer, squamous cell carcinoma of the head and neck (SCCHN), melanoma, non–small-cell lung cancer (NSCLC), or urothelial cancer. The primary objective of phase Ib was to determine the maximum tolerated dose (MTD) for lenvatinib plus pembrolizumab (200 mg intravenously every 3 weeks). In the preplanned phase II cohort expansion, the primary objective was objective response rate at week 24 (ORR week 24 ) at the recommended phase II dose. RESULTS Overall, 137 patients were enrolled during phase Ib (n = 13) and the initial phase II expansion (n = 124). Two dose-limiting toxicities (DLTs; grade 3 arthralgia and grade 3 fatigue) were reported in the initial dose level (lenvatinib 24 mg/d plus pembrolizumab). No DLTs were observed in the subsequent dose–de-escalation cohort, establishing the MTD and recommended phase II dose at lenvatinib 20 mg/d plus pembrolizumab. ORR week24 was as follows: RCC, 63% (19/30; 95% CI, 43.9% to 80.1%); endometrial cancer, 52% (12/23; 95% CI, 30.6% to 73.2%); melanoma, 48% (10/21; 95% CI, 25.7% to 70.2%); SCCHN, 36% (8/22; 95% CI, 17.2% to 59.3%); NSCLC, 33% (7/21; 95% CI, 14.6% to 57.0%); and urothelial cancer 25% (5/20; 95% CI, 8.7% to 49.1%). The most common treatment-related adverse events were fatigue (58%), diarrhea (52%), hypertension (47%), and hypothyroidism (42%). CONCLUSION Lenvatinib plus pembrolizumab demonstrated a manageable safety profile and promising antitumor activity in patients with selected solid tumor types.
Article
Full-text available
Purpose Anlotinib is a novel multi-target tyrosine kinase inhibitor (TKI) for tumor angiogenesis and tumor cell proliferation. The efficacy of anlotinib as a third-line or beyond therapy for SCLC was confirmed in the ALTER1202 trial. For lung cancer patients treated with antiangiogenesis agents, the phenomenon of cavitation is commonly seen in the lung target lesions. The impact of tumor cavitation on survival in lung cancer patients treated with vascular-targeted therapy remains controversial. Our retrospective study was to investigate the prognostic value of tumor cavitation in extensive-stage SCLC patients treated with anlotinib. Methods A total of 73 extensive-stage SCLC patients confirmed by histopathology from January 2018 to January 2019 were retrospectively analyzed. All patients received anlotinib therapy at Shanghai Chest Hospital. We defined tumor cavitation of the lung target lesions as that part of solid component was changed to air-filled area according to chest CT. Progression-free survival (PFS) was calculated from the beginning of anlotinib therapy to the disease progression or the last follow-up visit. Results Eleven (15.0%) patients had tumor cavitation during anlotinib therapy. The ORR of the 73 patients was 15.1%. Multivariate logistic regression analysis showed that tumor cavitation during anlotinib therapy was not associated with gender (P = 0.630), age (P = 0.190), smoking status (P = 0.165), anatomy type (P = 0.641), and the line of anlotinib therapy (P = 0.302). The median PFS of all patients was 2.6 months (95%CI 2.1–3.2). According to the univariate analysis, the median PFS in patients with and without tumor cavitation was 5.0 months and 2.2 months, respectively, and the difference was statistically significant (P = 0.041). According to the multivariate analysis, tumor cavitation was an independent factor for PFS after adjusting gender, age, smoking status, anatomy type, the line of anlotinib therapy, tumor cavitation, and response to anlotinib (adjusted HR 0.316, 95%CI 0.142–0.702; P = 0.005). Conclusions In 73 extensive-stage SCLC patients treated with anlotinib, no demographic/clinical characteristic was related to tumor cavitation, and tumor cavitation was an independent factor in predicting better PFS.
Article
Background We did a phase 2 trial of pembrolizumab in patients with non-small-cell lung cancer (NSCLC) or melanoma with untreated brain metastases to determine the activity of PD-1 blockade in the CNS. Interim results were previously published, and we now report an updated analysis of the full NSCLC cohort. Methods This was an open-label, phase 2 study of patients from the Yale Cancer Center (CT, USA). Eligible patients were at least 18 years of age with stage IV NSCLC with at least one brain metastasis 5–20 mm in size, not previously treated or progressing after previous radiotherapy, no neurological symptoms or corticosteroid requirement, and Eastern Cooperative Oncology Group performance status less than two. Modified Response Evaluation Criteria in Solid Tumors (mRECIST) criteria was used to evaluate CNS disease; systemic disease was not required for participation. Patients were treated with pembrolizumab 10 mg/kg intravenously every 2 weeks. Patients were in two cohorts: cohort 1 was for those with PD-L1 expression of at least 1% and cohort 2 was patients with PD-L1 less than 1% or unevaluable. The primary endpoint was the proportion of patients achieving a brain metastasis response (partial response or complete response, according to mRECIST). All treated patients were analysed for response and safety endpoints. This study is closed to accrual and is registered with ClinicalTrials.gov, NCT02085070. Findings Between March 31, 2014, and May 21, 2018, 42 patients were treated. Median follow-up was 8·3 months (IQR 4·5–26·2). 11 (29·7% [95% CI 15·9–47·0]) of 37 patients in cohort 1 had a brain metastasis response. There were no responses in cohort 2. Grade 3–4 adverse events related to treatment included two patients with pneumonitis, and one each with constitutional symptoms, colitis, adrenal insufficiency, hyperglycaemia, and hypokalaemia. Treatment-related serious adverse events occurred in six (14%) of 42 patients and were pneumonitis (n=2), acute kidney injury, colitis, hypokalaemia, and adrenal insufficiency (n=1 each). There were no treatment-related deaths. Interpretation Pembrolizumab has activity in brain metastases from NSCLC with PD-L1 expression at least 1% and is safe in selected patients with untreated brain metastases. Further investigation of immunotherapy in patients with CNS disease from NSCLC is warranted. Funding Merck and the Yale Cancer Center.
Article
Background LEN is a multikinase inhibitor of VEGFR 1–3, FGFR 1–4, PDGFRα, RET, and KIT. PEMBRO is an anti-PD-1 monoclonal antibody. LEN monotherapy is approved for first-line treatment of uHCC. PEMBRO monotherapy has shown significant efficacy in the second-line treatment of HCC. We report updated results from a phase 1b trial of LEN + PEMBRO in uHCC. Methods In this open-label, phase 1b study of LEN + PEMBRO in uHCC, 104 patients (pts) with BCLC stage B (not amenable for transarterial chemoembolization) or C, Child-Pugh class A, and ECOG PS ≤ 1 received LEN (body wt ≥ 60 kg: 12 mg/day; <60 kg: 8 mg/day QD) and PEMBRO (200 mg IV Q3W). Primary endpoints were safety (DLT part) and efficacy (expansion part; objective response rate [ORR] and duration of response [DOR]). Investigators performed tumor assessments using modified RECIST for HCC. No dose-limiting toxicities (DLTs) were reported in the DLT part (n = 6). Enrollment was expanded to approximately 100 pts (expansion part). Recruitment (N = 104) was completed April 2019. We present results for the first 67 pts enrolled by December 31, 2018. Results Pts received LEN + PEMBRO (DLT-evaluation part, n = 6; expansion part, n = 61). At data cutoff (June 30, 2019), 34 (50.7%) pts remained on study treatment; median duration of follow-up was 11.7 months (95% CI, 7.8-17.6). Serious adverse events occurred in 42 (62.7%) pts; no new safety signals emerged. Updated safety data will be provided. ORR (including unconfirmed responses) was 44.8% (Table) and compared favorably with LEN arm of REFLECT trial (24.1%; Kudo M. Lancet. 2018). Median DOR was 18.7 months (95% CI, 6.9-NE). Conclusions LEN + PEMBRO showed promising antitumor activity and an acceptable safety profile in pts with uHCC. Blinded independent central review is in progress. A phase 3 trial (LEAP-002; NCT03713593) is ongoing to assess LEN + PEMBRO vs LEN alone as first-line therapy for pts with uHCC. Table:747PResponse Parameter, n (%)LEN + PEMBRO (n = 67)mRECIST Per InvestigatorORR (CR + PR + unconfirmed PR or CR)a Best Overall ResponseCR.PRaStable diseaseProgressive diseaseUnknown/not evaluable30 (44.8)4 (6.0)26 (38.8)25 (37.3)6 (9.0)6 (9.0)aIncludes unconfirmed partial responses (2 patients). CR, complete response; (m)RECIST, (modified) Response Evaluation Criteria In Solid Tumors [Lencioni et al. Semin Liver Dis. 2010;30:52-60]; ORR, objective response rate; PR, partial response. Clinical trial identification NCT03006926; Release date: December 30, 2016. Editorial acknowledgement Medical writing support was provided by April Suriano, PhD, of Oxford PharmaGenesis, Newtown, PA, USA and was funded by Eisai Inc. Legal entity responsible for the study Eisai Inc. Funding Eisai Inc. and Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Disclosure J. Llovet: Advisory / Consultancy, Research grant / Funding (self): Bayer; Advisory / Consultancy, Research grant / Funding (self): Eisai; Advisory / Consultancy, Research grant / Funding (self): Bristol-Myers Squibb; Advisory / Consultancy, Research grant / Funding (self): Ipsen; Advisory / Consultancy: Eli Lilly; Advisory / Consultancy: Celsion Corporation; Advisory / Consultancy: Exelixis; Advisory / Consultancy: Merck; Advisory / Consultancy: Glycotest; Advisory / Consultancy: Navigant; Advisory / Consultancy: Leerink Swann LLC; Advisory / Consultancy: Midatech Ltd; Advisory / Consultancy: Fortess Biotech; Advisory / Consultancy: Sprink Pharmaceuticals; Advisory / Consultancy: Nucleix; Advisory / Consultancy: Can-Fite Biopharma. K.V. Shepard: Full / Part-time employment: Eisai Inc. R.S. Finn: Advisory / Consultancy, Research grant / Funding (institution): Bayer; Advisory / Consultancy, Research grant / Funding (institution): Bristol-Myers Squibb; Advisory / Consultancy, Research grant / Funding (institution): Eisai; Advisory / Consultancy: Genentech/Roche; Advisory / Consultancy, Research grant / Funding (institution): Lilly; Advisory / Consultancy, Research grant / Funding (institution): Merck; Advisory / Consultancy, Research grant / Funding (institution): Novartis; Advisory / Consultancy, Research grant / Funding (institution): Pfizer. M. Ikeda: Honoraria (self): Abbott, Bayer, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, Eisai, Lilly, Nobelpharma, Novartis, Otsuka, Taiho Pharmaceutical, Teijin Pharma, Yakult Honsha; Advisory / Consultancy: Bayer, Daiichi Sankyo, Eisai, Kyowa Hakko Kirin, MSD, NanoCarrier, Novartis, Shire, Teijin Pharma, Lilly; Research grant / Funding (self): ASLAN Pharmaceuticals, AstraZeneca, Baxalta/Shire, Bayer, Bristol-Myers Squibb, Chugai Pharma, Eisai, Kowa, Kyowa Hakko Kirin, Lilly, Merck Serono, MSD, NanoCarrier, Novartis, Ono Pharmaceutical, Taiho Pharmaceutical, Takara Bio, Yakult Honsha. M. Sung: Advisory / Consultancy: Bayer; Advisory / Consultancy: Eisai; Advisory / Consultancy: Exelixis. A.D. Baron: Speaker Bureau / Expert testimony: Amgen; Speaker Bureau / Expert testimony: Bristol-Myers Squibb; Speaker Bureau / Expert testimony: Genentech/Roche; Speaker Bureau / Expert testimony: Lilly; Speaker Bureau / Expert testimony: Merck. M. Kudo: Honoraria (self), Research grant / Funding (self): AbbVie; Honoraria (self), Advisory / Consultancy: Bayer; Honoraria (self), Advisory / Consultancy, Research grant / Funding (self): Bristol-Myers Squibb; Honoraria (self): EA Pharma; Honoraria (self), Advisory / Consultancy: Eisai; Honoraria (self): Gilead Sciences; Honoraria (self): Merck Serono; Honoraria (self), Advisory / Consultancy: MSD; Honoraria (self): Novartis; Honoraria (self): Pfizer; Honoraria (self), Research grant / Funding (self): Taiho Pharmaceutical; Research grant / Funding (self): Astellas Pharma; Research grant / Funding (self): Chugai Pharma; Research grant / Funding (self): Daiichi Sankyo; Research grant / Funding (self): Otsuka. T. Okusaka: Honoraria (self): AstraZeneca, Bayer, Bristol-Myers Squibb, Celgene, Chugai Pharma, Daiichi Sankyo, EA Pharma, Eisai, Fujifilm, Lilly Japan, Nippon Chemiphar, Nobelpharma, Novartis, Ono Pharmaceutical, Pfizer, Sumitomo Dainippon, Taiho Pharmaceutical, Teijin Pharma, Yakult; Advisory / Consultancy: Daiichi Sankyo, Sumitomo Dainippon, Taiho Pharmaceutical, Zeria Pharmaceutical; research funding from AstraZeneca, Baxter, Bayer, Chugai Pharma, Dainippon Sumitomo Pharma, Eisai, Kowa, Kyowa Hakko Kirin, Lilly Japan, Merck Serono, NanoCarrier, Nippon Boeh. M. Kobayashi: Speaker Bureau / Expert testimony: Eisai Inc. H. Kumada: Honoraria (self): AbbVie; Honoraria (self): Bristol-Myers Squibb; Honoraria (self): Gilead Sciences; Honoraria (self): MSD; Honoraria (self): Sumitomo; Honoraria (self): Dainippon. S. Kaneko: Honoraria (self): EA Pharma, MSD, Aska Pharma, Astellas Pharma, AbbVie, Eisai, Eidia, Otsuka, Gilead Sciences, Sysmex Corporation, Daiichi Sankyo, Taisho Toyama, Sumitomo Dainippon, Taiho, Takeda, Chugai, Bayer, Bristol-Myers, Mylan EPD, Miyarisan, Mochida, Kowa, Mitsubis; Research grant / Funding (self): Gilead Sciences, AbbVie, Daiichi Sankyo, Astellas Pharma, Takeda, Nippon Boehringer Ingelheim, MSD, Ono Pharma, Pfizer, Shionogi, Teijin Pharma, Mitsubishi Tanabe, Zeria, Abbott Vascular Japan, Taisho Toyama, Novartis, Terumo Corporation, Taiho, Novo Nord. K. Mamontov: Honoraria (self): Bayer; Honoraria (self): Eisai; Honoraria (self): Merck; Honoraria (self): MSD; Honoraria (self): Medtronic; Honoraria (self): Roche. T. Meyer: Advisory / Consultancy: Bristol-Myers Squibb; Advisory / Consultancy: Bayer; Advisory / Consultancy: Eisai; Advisory / Consultancy: BTG; Advisory / Consultancy: AstraZeneca; Advisory / Consultancy: BeiGene; Advisory / Consultancy: Tarveda; Advisory / Consultancy: MSD. K. Mody: Full / Part-time employment: Eisai Inc. T. Kubota: Full / Part-time employment: Eisai Co., Ltd. K. Saito: Full / Part-time employment: Eisai Inc. A.B. Siegel: Full / Part-time employment: Merck. L. Dubrovsky: Full / Part-time employment: Merck. A.X. Zhu: Advisory / Consultancy: AstraZeneca; Advisory / Consultancy, Research grant / Funding (institution): Bayer; Advisory / Consultancy, Research grant / Funding (institution): Bristol-Myers Squibb; Advisory / Consultancy: Eisai; Advisory / Consultancy: Exelixis; Advisory / Consultancy, Research grant / Funding (institution): Merck; Advisory / Consultancy, Research grant / Funding (institution): Lilly; Advisory / Consultancy, Research grant / Funding (institution): Novartis; Advisory / Consultancy: Sanofi. All other authors have declared no conflicts of interest.