17
Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR–MET Bispecific Antibody, in Diverse Models of EGFR Exon 20 Insertion–Driven NSCLC Jiyeon Yun 1 , Soo-Hwan Lee 2 , Seok-Young Kim 1 , Seo-Yoon Jeong 1 , Jae-Hwan Kim 1 , Kyoung-Ho Pyo 1 , Chae-Won Park 1 , Seong Gu Heo 1 , Mi Ran Yun 2 , Sangbin Lim 1 , Sun Min Lim 3 , Min Hee Hong 3 , Hye Ryun Kim 3 , Meena Thayu 4 , Joshua C. Curtin 4 , Roland E. Knoblauch 4 , Matthew V. Lorenzi 4 , Amy Roshak 4 , and Byoung Chul Cho 3 RESEARCH ARTICLE Cancer Research. on January 24, 2021. © 2020 American Association for cancerdiscovery.aacrjournals.org Downloaded from Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR–MET Bispecific Antibody, in Diverse Models of EGFR Exon 20 Insertion–Driven NSCLC Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park1, Seong Gu Heo1, Mi Ran Yun2, Sangbin Lim1, Sun Min Lim3, Min Hee Hong3, Hye Ryun Kim3, Meena Thayu4, Joshua C. Curtin4, Roland E. Knoblauch4, Matthew V. Lorenzi4, Amy Roshak4, and Byoung Chul Cho3

ReseaRch aRticle

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 2: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

AUGUST 2020 CANCER DISCOVERY | 1195

1 Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of South Korea. 2 JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, Republic of South Korea. 3 Division of Medical Oncol-ogy, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea. 4 Janssen Research and Development, Spring House, Pennsylvania. Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/). Corresponding Author: Byoung Chul Cho, Yonsei University College of Medicine, 51 Yonseiro, Seodaemun-gu, Seoul 120-752, Republic of South Korea. Phone: 822-2228-8126; Fax: 822-393-3652; E-mail: [email protected] Cancer Discov 2020;10:1194–209 doi: 10.1158/2159-8290.CD-20-0116 ©2020 American Association for Cancer Research.

aBstRact EGFR exon 20 insertion driver mutations (Exon20ins) in non–small cell lung cancer (NSCLC) are insensitive to EGFR tyrosine kinase inhibitors (TKI). Amivantamab

(JNJ-61186372), a bispecifi c antibody targeting EGFR–MET, has shown preclinical activity in TKI-sensitive EGFR -mutated NSCLC models and in an ongoing fi rst-in-human study in patients with advanced NSCLC. However, the activity of amivantamab in Exon20ins-driven tumors has not yet been described. Ba/F3 cells and patient-derived cells/organoids/xenograft models harboring diverse Exon20ins were used to characterize the antitumor mechanism of amivantamab. Amivantamab inhibited proliferation by effectively downmodulating EGFR–MET levels and inducing immune-directed antitumor activity with increased IFN γ secretion in various models. Importantly, in vivo effi cacy of amivantamab was superior to cetuximab or poziotinib, an experimental Exon20ins-targeted TKI. Amivantamab produced robust tumor responses in two Exon20ins patients, highlighting the important translational nature of this pre-clinical work. These fi ndings provide mechanistic insight into the activity of amivantamab and support its continued clinical development in Exon20ins patients, an area of high unmet medical need.

SIGNIFICANCE: Currently, there are no approved targeted therapies for EGFR Exon20ins–driven NSCLC. Preclinical data shown here, together with promising clinical activity in an ongoing phase I study, strongly support further clinical investigation of amivantamab in EGFR Exon20ins–driven NSCLC.

intRoDuction Molecular segmentation of advanced non–small cell lung

cancer (NSCLC) based on oncogenic driver mutations has improved the overall survival and quality of life for patients with actionable driver mutations, and solidifi ed solid-tumor targeted therapy. Mutations in the EGFR ( 1, 2 ) gene constitu-tively activate downstream growth and survival signaling path-ways leading to dependency on the EGFR pathway for tumor growth. Nearly 20% of Caucasians and up to 50% of Asians with lung adenocarcinomas harbor mutations in EGFR ( 3, 4 ).

EGFR activating mutations have been reported in the fi rst four exons (18 through 21) of its tyrosine kinase domain. NSCLCs that harbor “classic” EGFR mutations in exons 18, 19, and 21, for example, exon 19 deletions or L858R, are sen-sitive to treatment with fi rst-, second-, and third-generation EGFR tyrosine kinase inhibitors (TKI) such as erlotinib, afatinib, and osimertinib ( 5, 6 ). In contrast, the EGFR exon 20

mutations encompass nucleotides that translate into amino acids at position 762–823, and include a C-helix (762–766) followed by a loop (position 767–775; ref. 7 ). The insertion mutations of one to seven amino acids in exon 20 form a wedge at the end of the C-helix in EGFR that promotes active kinase conformation. EGFR exon 20 insertion driver muta-tions (Exon20ins), a distinct and highly heterogeneous sub-set of NSCLCs, represent 4% to 12% of all EGFR mutations ( 7, 8 ). These Exon20ins mutations are generally insensitive to approved EGFR TKIs and are associated with poor prognosis, thus representing an area of high unmet medical needs ( 6, 9 ).

Recently, poziotinib and TAK-788 have been undergoing clinical evaluation in patients whose tumors carry EGFRExon20ins mutations ( 10, 11 ). Despite initial promising effi -cacy, the Zenith 20 trial demonstrated that poziotinib had a low response rate (RR; ∼14%) in patients with NSCLC with the EGFR Exon20ins mutation. Furthermore, both pozi-otinib and TAK-788 have high rates of EGFR wild-type (WT)–driven toxicity due to the lack of selectivity upon Exon20ins as compared with EGFR WT and other kinases, limiting their clinical utility ( 6 ).

Amivantamab (JNJ-61186372; Fig.  1A ) is an EGFR–MET bispecifi c antibody with immune cell–directing activity that targets activating and resistant EGFR mutations and METmutations and amplifi cations. Ongoing fi rst-in-human studies in patients with advanced, refractory EGFR -mutant NSCLC have demonstrated preliminary clinical activity of amivantamab in patients with diverse EGFR mutations ( 12, 13 ). Of note, amivantamab showed promising effi cacy (30% RR) with a manageable safety profi le in patients with heavily pretreated EGFR Exon20ins NSCLC. Although amivantamab has been reported to harbor activity in preclinical tumor models driven by EGFR mutations sensitive to approved TKIs (e.g., L858R and Exon 19 deletions; ref. 14 ), its activity has not yet been explored in the context of EGFR Exon20ins.

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 3: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1196 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

Here, we comprehensively evaluated the antitumor activity and mechanisms of action (MOA) of amivantamab in multi-ple EGFR Exon20ins models, including engineered cell lines, patient-derived cells (PDC), and patient-derived xenografts (PDX). We also present evidence of clinical activity in two case studies of patients with EGFR Exon20ins NSCLC treated with amivantamab from an ongoing phase I clinical trial, highlighting the important translational nature of this work.

ResultsAmivantamab Inhibits Proliferation of Ba/F3 Cells Harboring Diverse EGFR Exon20ins Mutations

To demonstrate the antitumor activity of amivantamab in the context of Exon20ins, multiple Exon20ins were sta-bly expressed in Ba/F3 cells. Five distinct Exon20ins were introduced (Fig.  1B), all of which have been observed in patients with NSCLC (V769_D770insASV, D770delinsGY, H773_V774insH, Y764_V765insHH, and D770_N771ins-SVD; refs. 15, 16). In Ba/F3 cells treated with amivantamab ranging from 0.05 to 1 mg/mL, a significant and dose-dependent decrease in Ba/F3 cell viability (P < 0.0001) was observed in all five EGFR Exon20ins mutations (Fig. 1C). In contrast, treatment with the first- and third-generation irre-versible EGFR TKIs gefitinib and osimertinib, respectively, showed limited antiproliferative activity compared with ami-vantamab (Fig.  1C), confirming the well-known resistance of Exon20ins to EGFR TKIs. No effect on cell viability was observed when IgG1 control antibodies were used in the same Ba/F3 cell lines (Supplementary Fig.  S1A). In tumor mod-els driven by TKI-sensitive EGFR mutations such as L858R or Exon 19 deletions, amivantamab has several proposed MOAs including blocking ligand binding, receptor down-modulation, downstream signaling inhibition, and trigger-ing immune-directed antitumor activity (17). To determine whether these MOAs are also observed in the context of Exon20ins and contribute to the observed antiproliferative activity, immunoblot analysis was performed (Fig.  1C) in Ba/F3 cells overexpressing the EGFR D770delinsGY and H773_V774insH Exon20ins mutations. The total EGFR lev-els were reduced following treatment with amivantamab, compared with those of untreated cells (Fig. 1D; Supplemen-tary Fig. S1B) or cells treated with the IgG1 control antibody (Supplementary Fig.  S1C). Consistent with the reduction in EGFR expression levels, the EGFR downstream signaling pathways phospho-EGFR (pEGFR), phospho-AKT (pAKT), phosho-ERK (pERK), and phospho-S6 (pS6) were also signifi-cantly reduced following amivantamab treatment (Fig. 1D), suggesting that amivantamab targeted EGFR and inhibited EGFR-related downstream signaling cascades. Similar results were observed in Ba/F3 cells expressing the V769insASV, Y764 insHH, and D770_N771insSVD Exon20ins mutations (Supplementary Fig. S1B). Although 100 nmol/L of gefitinib and osimertinib reduced pEGFR in Ba/F3 cells overexpress-ing D770delinsGY and H773_V774insH, downstream EGFR signaling pathway components were not significantly inhib-ited, which correlated with the lack of TKI effects on cell viability (Fig. 1E; Supplementary Fig. S1D). In recent studies, poziotinib has shown antitumor activity in EGFR Exon20ins NSCLC (18, 19). We further assessed the cell viability test for

poziotinib in Ba/F3 cells overexpressing EGFR Exon20ins (Supplementary Table S1). Consistent with a previous report (18), poziotinib strongly inhibited the cell viability in the mutant EGFR Exon20ins cells (IC50 ranging from 0.8 to 10.9 nmol/L). As reported in a previous study (20), poziotinib also potently suppressed proliferation of Ba/F3 cells harboring WT EGFR (IC50 = 0.8 nmol/L). To present the selectivity for Exon20ins mutation in a more balanced manner, we com-pared antiproliferative potency between amivantamab and poziotinib in EGFR Exon20ins mutants over WT EGFR. Pozi-otinib exhibited lower EGFR Exon20ins–mutant selectivity over WT EGFR compared with amivantamab, suggesting that poziotinib may adversely affect normal tissues, thereby pro-ducing substantial toxicities such skin rash and diarrhea (21). To better understand the mechanisms involved in amivan-tamab-mediated cellular cytotoxicity, we assessed the effect of amivantamab treatment on cell-cycle progression and programmed cell death. In Ba/F3 cells expressing the EGFR D770delinsGY and H773_V774insH Exon20ins mutations, an accumulation of cells in G1 phase was observed in ami-vantamab-treated cells compared with vehicle-treated cells (Fig. 1F). As EGFR TKIs have been reported to drive apoptosis in NSCLC cells harboring sensitizing EGFR mutations (22, 23), we investigated whether treatment with amivantamab resulted in engagement of the apoptotic machinery. Amivan-tamab treatment resulted in the induction of proapoptotic proteins, including BIM and cleaved caspase-3 (Fig. 1G), sug-gesting that amivantamab, in addition to inhibiting down-stream EGFR signaling cascade, also induced apoptosis in a BIM- and caspase-dependent manner.

Amivantamab Displays Antitumor Activity in PDCs and Organoids

To extend our findings from Ba/F3 cells engineered to express the exogenous EGFR Exon20ins mutations, we evalu-ated the activity of amivantamab in several PDCs harboring the Exon20ins. The antitumor activity of amivantamab and associated mechanistic endpoints were evaluated in PDCs generated from patients harboring P772ins_H773insPNP (DFCI-127), H773_V774insNPH (DFCI-58), and S768_D770dup (YU-1163) Exon20ins mutations (Supplementary Fig.  S2A–S2C; Supplementary Table S2). In both DFCI-127 and DFCI-58 cells, amivantamab treatment resulted in decreased expression of total EGFR and MET levels, as well as inhibition of pEGFR, pMET, pAKT, pERK, and pS6 (Fig.  2A), consistent with the results observed in Ba/F3 cell lines harboring EGFR Exon20ins mutations. Analysis of cell viability and colony formation revealed that amivantamab dose-dependently inhibited the cell growth and proliferation of PDCs, compared with IgG1 controls (Fig.  2B and C). In contrast to the significant reduction in EGFR, MET, pEGFR, pMET, pAKT, and pS6 in DFCI-127 and DFCI-58 cells, YU-1163 treated with amivantamab unexpectedly revealed an induction of pERK (Fig. 2A). Consistent with this result, the growth of YU-1163 was not inhibited after amivantamab treatment for 72 hours or following long-term treatment (Fig.  2B and C). From the whole-exome sequencing data of YU-1163, we observed a co-occurring mutation in the TP53 gene (R280T; 96% of mutant allele frequency; Supplementary Fig. S2C and S2D). According to recent studies, mutations

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 4: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1197

Figure 1.  Amivantamab shows antitumor activity and suppresses EGFR and MET signaling pathways in Ba/F3 cells with EGFR Exon20ins mutations. A, Schematic of the structure of amivantamab, an EGFR and MET bispecific antibody. B, Schematic of EGFR Exon20 insertions in stable Ba/F3 cells, PDC, PDO, and PDX models. C, The viability of Ba/F3 cells was determined via CellTiter-Glo. Amivantamab, gefitinib, or osimertinib were treated for 72 hours. Data are presented as averages ± SD of triplicate independent experiments. *, P < 0.0001; **, P < 0.001; Student t test. D, Ba/F3 cells overexpressing the indicated EGFR Exon20ins mutations were treated with amivantamab for 72 hours at the indicated concentrations. E, Ba/F3 cells overexpressing the indicated EGFR Exon20ins mutations were treated with osimertinib or gefitinib for 6 hours at the indicated concentrations. Immu-noblot analysis was performed for EGFR, MET, AKT, ERK, and S6 expression after amivantamab treatment. Amivantamab inhibited the cell cycle and induced synergistic apoptosis in Ba/F3 cells overexpressing the EGFR Exon20ins mutations. F, Amivantamab induced G1-phase arrest after amivantamab treatment for 72 hours in Ba/F3 cells overexpressing the indicated types of EGFR Exon20ins mutations. Cell cycles were analyzed using propidium iodide staining and FACS analysis. Data are presented as averages ± SD of triplicate independent experiments. *, P < 0.0001; Student t test. G, BIM- and caspase-dependent apoptosis were induced during amivantamab treatment. The expression of BIM and cleaved caspase-3 were detected by Western blotting.

A

C V769insASV

* * * **

**

****

* *

* * * * * *

*

**

D770delinsGY

D770delinsGY

H773_V774insH

D770delinsGY

H773_V774insH

Amivantamab(mg/mL)

Vehicle

Cleaved caspase-3

GAPDH

p21

p27

p53

BIMEL

BIML

BIMs

0.1 mg/mL

1 mg/mL

Amivantamab

Amivantamab

Sub G1phase

G1phase

Sphase

G2–M phase

Sub G1phase

G1phase

Sphase

G2–M phase

* ** *

GAPDH

S6

pS6

ERK

pERK

AKT

pAKT

pEGFR

EGFR

GAPDH

0

10

20

30

40

Cel

l pop

ulat

ion

(%) 50

60

0

20

40

60

Cel

l pop

ulat

ion

(%)

80

S6

pS6

ERK

pERK

AKT

pAKT

pEGFR

EGFR

GAPDH

S6

pS6

ERK

pERK

AKT

pAKT

pEGFR

EGFR

Osimertinib (nmol/L) Gefitinib (nmol/L)0

0 10 100 10 100 0

0 0.1 1 0 0.1 1

10 100 10 1000.1 1 0 0.1

Amivantamab(mg/mL)1

MET

K409R F405L

ControlledFab-arm exchange

EGFR

+

Amivantamab

Extracellulardomain

Transmembranedomain Tyrosine kinase domain

S768dupS768dup

EGFR Ex20(762–823)

E762

A763

Y764

V765

M766

A767

A767dup D770insG

Y764insHH

V769insASV

H773insHD770insSVDD770delinsGY Ba/F3

S768

V769

D770

N771

P772

H773

V774

C775

P772insPNP

P773insNPH

PDCPDOPDX

I/II III/IV Ex18 Ex19 Ex20 Ex21 Ex22 Ex23

K409RF405L

Stable chinge

Matched CH3 domains

D770delinsGY H773ins H Y764insHH D770insSVD

0

Amivantamab(mg/mL)

Gefitinib(nmol/L)

Osimertinib(nmol/L)

Amivantamab(mg/mL)

Gefitinib(nmol/L)

Osimertinib(nmol/L)

Amivantamab(mg/mL)

Gefitinib(nmol/L)

Osimertinib(nmol/L)

Amivantamab(mg/mL)

Gefitinib(nmol/L)

Osimertinib(nmol/L)

Amivantamab(mg/mL)

Gefitinib(nmol/L)

Osimertinib(nmol/L)

00.

05 0.1

0.5 1 0

0.05 0.

10.

5 10 1 10 100 0 1 10 100 0 1 10 100 0

0.05 0.

10.

5 1 0 1 10 100 0

0.05 0.

10.

5 1 00.

05 0.1

0.5 10 1 10 100 0 1 10 100 0 1 10 100 0 1 10 1000 1 10 1000 1 10 100

Cel

l via

bilit

y (%

)

102030405060708090

100110120130

0102030405060708090

100110120130

0102030405060708090

100110120130

0102030405060708090

100110120130

0102030405060708090

100110120130

D

F G

E

B

H773_V774insH

D770delinsGY H773_V774insH

Osimertinib (nmol/L) Gefitinib (nmol/L)

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 5: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1198 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

Figure 2.  Amivantamab has antitumoral activity and suppresses EGFR and MET signaling pathways in PDCs and PDOs harboring EGFR Exon20ins mutations. A, PDCs with the indicated EGFR Exon20ins mutations were treated with amivantamab for 72 hours at the indicated concentrations. Immu-noblot analysis was performed for EGFR, MET, AKT, ERK, and S6 expression after amivantamab treatment. B, The viability of PDCs was determined via CellTiter-Glo. Amivantamab was treated for 72 hours. Data are presented as averages ± SD of triplicate independent experiments. *, P < 0.0001; **, P < 0.001; Student t test. C, Effects of amivantamab on the colony formation and cell proliferation of PDCs. Representative images and quantitative analysis of the colony formation assay. Data are presented as averages ± SD of triplicate independent experiments. *, P < 0.0001; Student t test. O.D., optical density. D and E, Dose–response curves of (D) YUO-036 (A767_V769dup) and (E) YUO-029 (S768_D770dup) PDOs treated with IgG1 control or amivantamab. Cell viability was measured using CellTiter-Glo 3D cell viability reagent 72 hours after drug treatment. Representative images of PDOs treated with amivantamab for 72 hours at the indicated concentrations. Data are presented as averages ± SD of triplicate independent experiments. *, P < 0.0001; Student t test; NS, not significant.

pEGFR

0 0.1 1 0 0.1 1 0 0.1 1Amivantamab(mg/mL)

DFCI-127(P772_H773insPNP)

A

B

C

D E

DFCI-58(H773_V774insNPH)

YU-1163(S768_D770dup)

pMET

MET

pAKT

pERK

ERK

pS6

S6

GAPDH

120100806040200

120

0

0.1

1

Amivantamab(mg/mL)

0

0.1

1

Amivantamab(mg/mL)

100

80

60

40

20

00 0.05 0.1 0.5 1

120100806040200

140

100120

806040200

0

0Amivantamab(mg/mL)

0.1 1 1 10.100.10

10 0.1

**

DFCI-127120100

O.D

(%

)

806040200

10 0.1

**

DFCI-58120100

O.D

(%

)

806040200

10 0.1

YU-1163120100

O.D

(%

)

806040200

0.05 0.1 0.5

lgG1 (mg/mL)

**** *

* * **

Amivantamab (mg/mL)

Amivantamab (mg/mL)lgG1 control (mg/mL)

*

*

YUO-036(A767_V769dup)

YUO-029(S768_D770dup)

* *

lgG1 (mg/mL)Amivantamab (mg/mL)

lgG1 (mg/mL)Amivantamab (mg/mL)

1 0 0.05 0.1 0.5 1 0 0.05 0.1 0.5 1

Cel

l via

bilit

y (%

)

Cel

l via

bilit

y (%

)

120

100

80

60

40

20

00 0.05 0.1 0.5 1

Amivantamab (mg/mL)lgG1 control (mg/mL)

NS

Cel

l via

bilit

y (%

)

Cel

l via

bilit

y (%

)

Cel

l via

bilit

y (%

)

AKT

EGFR

pEGFR

pMET

MET

pAKT

pERK

ERK

pS6

S6

GAPDH

AKT

EGFR

pEGFR

pMET

MET

pAKT

pERK

ERK

pS6

S6

GAPDH

AKT

EGFR

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 6: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1199

in TP53 commonly occurred with EGFR mutations in NSCLC. Particularly, TP53 mutations in exon 8 in patients with NSCLC with EGFR mutations show lower responsiveness to EGFR TKIs and worse prognosis than patients with WT TP53 (24, 25). Indeed, accumulated studies have revealed that the R280T mutation in TP53 plays crucial roles in the proliferation and survival of cancer cells, and knockdown of the mutant TP53 causes G2-phase arrest and apoptosis in bladder cancer cells (26, 27). As shown in Supplementary Fig. S2E, depletion of mutant TP53 by three different TP53-directed siRNAs significantly inhibited cell proliferation with a reduction in activated ERK in YU-1163 pretreated with 1 mg/mL amivantamab. Given that mutant TP53 is associated with EGFR-TKI resistance (28) and the depleted mutant TP53 restored the sensitivity of amivantamab by downregulation of pERK, induction of pERK following amivantamab treatment in YU-1163 cells might be a key regulator of cell survival, potentially through the cross-talk between mutant TP53 and ERK signaling cascade (29, 30). In addition, we generated two patient-derived organoid (PDO) models from plural effu-sion of patients who had A767_V769dup (YUO-036) and S768_D770dup (YUO-029) to recapitulate the phenotypic and molecular landscape of the original NSCLC with EGFR Exon20ins (Supplementary Fig. S2F and S2G; Supplementary Table S2). YUO-029 was derived from the same patient from whom YU-1163 PDC (S768_D770dup) was derived. As shown in Fig. 2D, YUO-036 was sensitive to amivantamab in a dose-dependent manner, whereas YUO-029 derived from the same patient with YU-1163 showed no significant decrease in cell viability following amivantamab treatment compared with IgG1 control (Fig. 2E). Taken together, these results indicate that amivantamab has potent antitumor activity in NSCLC patient-derived cancer cells with EGFR Exon20ins mutations by downmodulation of EGFR and MET signaling pathways.

EGFR and MET Are Internalized in Response to Amivantamab

Treatment with amivantamab results in downmodulation of EGFR and MET, as observed in Ba/F3 cells (Fig.  1) and PDCs (Fig.  2). According to many studies, anti-EGFR mAb induces internalization of EGFR, leading to downregula-tion of its expression on the cell surface (31, 32). To investi-gate whether amivantamab directly binds to EGFR on cells with EGFR Exon20ins mutation, Ba/F3 cells overexpress-ing D770delinsGY or H773_V774insH were incubated with 0.1 mg/mL IgG1 control and 0.1 mg/mL amivantamab. FACS was used to measure the level of plasma membrane–bound EGFR. EGFR expression on the plasma membrane began to dwindle by almost 2-fold 30 minutes after amivantamab treatment. The percentage changes in median fluorescence intensity (MFI) of EGFR relative to IgG1 control–treated cells at 30 minutes were 56% and 68% in D770delinsGY and H773_V774insH, respectively, and subsequently remained at 40% EGFR expression relative to IgG1 control–treated cells 72 hours after amivantamab treatment (Fig. 3A). To explore the internalization of MET as well as EGFR on PDCs har-boring EGFR Exon20ins, DFCI-127 and DFCI-58 PDCs were treated with 0.1 mg/mL amivantamab and the plasma mem-brane–bound MET and EGFR were measured 72 hours after amivantamab treatment (Fig. 3B and C). The results showed

that amivantamab reduced EGFR and MET on PDCs com-pared with IgG1 control. Immunofluorescence (IF) staining was used to visualize the internalization of EGFR and MET following amivantamab treatment. Treatment with 0.1 mg/mL amivantamab for 72 hours led to the redistribution of EGFR and MET receptors into internal compartments, whereas IgG-treated cells showed no change in the staining pattern for EGFR or MET (Fig. 3D; Supplementary Fig. S3). Internalization and subsequent downregulation of EGFR and MET receptors by lysosomes could account for the decreased EGFR and MET protein levels observed in the immunoblot, FACS, and IF assays following amivantamab treatment. To determine whether lysosomal degradation was involved in downregulating EGFR protein levels, Ba/F3 cells overexpressing D770delinsGY and H773_V774insH were treated with amivantamab in the absence and presence of the autophagy inhibitor bafilomycin. Bafilomycin treatment inhibited the degradation of EGFR (Fig. 3E), suggesting that downmodulation of the total EGFR protein level following amivantamab treatment may involve lysosomal degradation of internalized cell-surface receptors. Taken together, these results suggest that treatment with amivantamab induces receptor internalization and may contribute to the observed antiproliferative effects of amivantamab by inhibiting EGFR- and MET-mediated signaling.

Amivantamab Inhibits EGFR Exon20ins Mutation–Driven Growth of Ba/F3 and PDC Models In Vivo

To determine whether amivantamab is active against EGFR Exon20ins–derived tumors in vivo, xenograft models were gen-erated using Ba/F3 cells overexpressing EGFR D770delinsGY and H773_V774insH Exon20ins mutations and PDCs (DFCI-127 and YU-1163) harboring P772insPNP and S768_D770dup EGFR Exon20ins mutations, respectively. Mice were treated with amivantamab, IgG1 control, or vehicle at 30 mg/kg twice per week intraperitoneally. Amivantamab-treated mice showed reduced tumor volumes compared with vehicle- or IgG1 control–treated mice in the Ba/F3 cells bearing NOD.Cg-Prkdcscid II2rgtm1Sug/Jic (NOG) mice models (Fig. 4A and B; Supplementary Fig. S4A and S4B). Inhibition of tumor growth occurred early and was sustained 15 days following treatment. As shown in Ba/F3 cells and PDCs in vitro (Figs. 1D and 2A), protein expression of EGFR, MET, pEGFR, and pMET were significantly reduced following amivantamab treatment in the Ba/F3-bearing NOG mice models (Fig.  4C; Supplementary Fig. S4C). Similarly, in the PDC xenograft models, amivantamab-treated mice showed a reduction in tumor volume compared with vehicle-treated mice (Fig.  4D–G), as well as a reduction in EGFR, MET, pEGFR, and pMET protein levels (Fig.  4H and I). Intriguingly, although amivantamab could not inhibit the proliferation of YU-1163 PDC in vitro (Fig. 2), a dramatic tumor regression was observed in YU-1163–bearing BALB/c nude mice after amivantamab treatment (Fig. 4F), suggesting that additional factors might contribute to the in vivo antitu-mor effect of amivantamab. As mentioned above, poziotinib is a targeted agent that has shown preliminary clinical activity in EGFR Exon20ins disease (18, 19). We compared the antitumor activity and safety of poziotinib with those of amivantamab in YU-1163 (S768_D770dup)–bearing BALB/c nude mice and Ba/F3 cells overexpressing D770_N771insSVD-bearing

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 7: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1200 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

Figure 3.  Amivantamab strongly promotes internalization of EGFR and MET in Ba/F3 and PDC cells expressing EGFR Exon20ins mutations. The expression of EGFR and MET on the cell surface were determined by FACS analysis. A, EGFR expression on the plasma membrane was detected in Ba/F3 cells overexpressing D770delinsGY and H773_V774insH at the indicated time. After 0.1 mg/mL IgG1 control (cont) or 0.1 mg/mL amivantamab treat-ment for 72 hours, PE-EGFR and FITC-MET expression on the plasma membrane was detected in (B) DFCI-127 (P772_H773insPNP) and (C) DFCI-58 (H773_V774insNPH) cells. D, Amivantamab induced redistribution of EGFR and MET in DFCI-127 PDCs. IF staining for EGFR (green) and MET (red) in a panel of DFCI-127 treated with 0.1 mg/mL IgG1 control or 0.1 mg/mL amivantamab for 72 hours. E, Pretreatment with the autophagy inhibitor bafilomycin (100 nmol/L) for 30 minutes rescued the decreased EGFR expression in 1 mg/mL amivantamab-treated Ba/F3 cell lines overexpressing D770delinsGY or H773_V774insH.

AD770

delinsGYH773_V774

insHDFCI-127

(P772_H773insPNP)

D770delinsGY

EGFR

GAPDH

Amivantamab (1 mg/mL)

Bafilomycin (nmol/L)

IgG

1(0

.1 m

g/m

L)A

miv

anta

mab

(0.1

mg/

mL)

Am

ivan

tam

ab(0

.1 m

g/m

L)Ig

G1

coun

t(0

.1 m

g/m

L)

DF

CI-

127

1.02

FIT

C-M

ET

FIT

C-M

ET

FIT

C-M

ET

93.5

1.89 3.63

81.9 7.32

9.19 1.56

12.7 19.3

47.1 20.9

6.69 35.5

27.4 30.4

DFCI-58(H773_V774insNPH)

00 103 104 105

100

30 m

inco

unt

200

300

00 103 104 105

100

6 h

coun

t

200

300

0

0

0

PE-EGFR

PE-EGFR

EGFR

IgG1 control (0.1 mg/mL)

Amivantamab (0.1 mg/mL)

% MFI ratio

D770delinsGY

30 min

6 h

24 h

48 h

72 h

55.9

58.9

47.5

45.7

44.4

68.0

49.1

49.3

46.9

42.9

H773_V774InsH

EGFR

EGFR MET DAPI Merge

PE-EGFR

PE-EGFR

102

103 104 105

0 103 104 105 0 103 104 105

103

104

0

102

103

104

0

102

103

104

FIT

C-M

ET

0

0

102

103 104 105

103

104

0 103 104 105

100

200

300

00 103 104 105

100

24 h

coun

t

200

300

00 103 104 105

100

200

300

00 103 104 105

100

48 h

coun

t

200

300

00 103 104 105

100

200

300

00 103 104 105

100

72 h

coun

t 200

300

00 103 104 105

100

200

300

400

00 103 104 105

100

200

300

B

D

E

C

H773_V774insH

+− +−− 100

+− +−− 100

NOG mice (Supplementary Fig.  S4D and S4E). Using the previously reported dosing regimen of 5 mg/kg poziotinib, once a day (18), sudden death occurred within 6 days of treatment. Skin toxicity analyses with poziotinib and amivan-tamab revealed that poziotinib-treated mice showed severe skin toxicities on the face, abdomen, and back at dose of 5 and 10 mg/kg, whereas 30 mg/kg amivantamab showed only mini-mal keratosis on the face (Supplementary Fig. S4F and S4G). In addition to skin toxicity, a dramatic loss of body weight was

observed in poziotinib-treated mice compared with amivan-tamab-treated mice (Supplementary Fig. S4H). The favorable toxicity profiles with amivantamab were consistent with those shown in an ongoing phase I study (13).

Amivantamab Induces Antibody-Dependent Cell-Mediated Cytotoxicity in Exon20ins Models

The process of antibody-dependent cell-mediated cytotox-icity (ADCC) is known to be initiated when both the target

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 8: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1201

Figure 4.  Amivantamab reduces tumor burden in Ba/F3 cells and PDCs with EGFR Exon20ins xenograft models. Antitumor effects of amivantamab in (A–C) Ba/F3 cells overexpressing D770delinsGY- or H773_V774insH-bearing NOG mice and (D–I) DFCI-127- or YU-1163–bearing NOG or BALB/c nude mice, respectively. (continued on next page)

800

A B C

700

600

500

400

300

200

100

0

0

100

200

300

400

500

600

700

800

900

1,000

800

pEGFR

pMET

MET

GAPDH

EGFR

700

600

500

400

300

200

100

0

0

100

200

300

400500

600

700

800

900

1,000

Vehicle

Vehicle

#1 #2 #2#1

Amivantamab30 mg/kg

Vehicle Amivantamab30 mg/kg

Amivantamab(30 mg/kg)

D770delinsGY

pEGFR

pMET

MET

GAPDH

EGFR

Vehicle

#1 #2 #2#1

Amivantamab(30 mg/kg)

H773_V774insH

D770delinsGY

H773_V774insH

D770delinsGY

H773_V774insH

1 3 5

Day after treatment

Tum

or v

olum

e (m

m3 )

Tum

or v

olum

e (m

m3 )

% c

hang

e in

tum

or v

olum

e%

cha

nge

in tu

mor

vol

ume

7 9 11

Vehicle

lgG1 30 mg/kgAmivantamab 30 mg/kg *

Vehicle

lgG1 30 mg/kg

Amivantamab 30 mg/kg*

13 15

1 3 5

Day after treatment

7 9 11 13 15

cell antigen and an activated Fcγ receptor are engaged, respec-tively, by the Fab and Fc portions of an antibody. The effector cells, mainly natural killer (NK) cells, trigger degranulation and subsequent cytokine production, resulting in the elimi-nation of the target cells (33). To determine whether ADCC plays a role in amivantamab-mediated antitumor activity, ADCC assays were performed using PDCs (DFCI-127 and YU-1163) expressing EGFR Exon20ins mutations cocultured with peripheral blood mononuclear cells (PBMC) as effector cells [effector:target (E:T) = 50:1]. Treatment with amivan-tamab resulted in cytotoxicity in both PDCs in a dose-depend-ent manner and to a greater extent than cetuximab, a mAb targeting EGFR (Fig.  5A–C). By extension, cetuximab treat-ment led to a less-pronounced reduction in tumor volume in YU-1163–bearing BALB/c nude mice models relative to that observed with amivantamab (Supplementary Fig. S5A). Ami-vantamab-mediated cellular cytotoxicity shown in Fig.  5A was significantly impaired by incubation with an Fc receptor (FcR) blocker in DFCI-127 and YU-1163 PDCs (Fig.  5D), suggesting that the amivantamab-mediated ADCC effect requires the interaction with FcRs on PBMCs. Similarly, the antitumor effect of amivantamab was abrogated in vivo when amivantamab was cotreated with anti-mouse CD16/CD32 antibodies to block FcRγIII/FcRγII on monocytes/mac-rophages and NK cells in YU-1163–bearing BALB/c nude mice (Supplementary Fig.  S5A). It is known that inflammatory cytokines such as IFNγ and TNFα are secreted from infected

monocytes and activated NK cells during ADCC, encourag-ing antigen presentation and adaptive immune responses (34, 35). To explore the correlation between amivantamab-dependent ADCC and secreted IFNγ levels, we measured the level of IFNγ in a medium cocultured with PDCs and PBMC after amivantamab treatment. Consistent with the degree of the ADCC effect, IFNγ levels were significantly increased with amivantamab treatment compared with cetuximab treatment (Fig. 5E). Treatment with a FcR blocker reduced IFNγ secre-tion, indicating that IFNγ secretion was dependent on the interaction between the Fc domain of amivantamab and the FcR on immune cells (Fig.  5F). Induced inflammatory cytokines including IFNγ secreted from NK cells activated by amivantamab bound to EGFR and MET on EGFR Exon20ins-driven tumors may lead to the recruitment and activation of adjacent immune cells to tumor cells in vivo. To explore this, we analyzed the infiltration of macrophages and NK cells into the tumor in a PDX model (YHIM-1029), which was generated from a patient-derived tumor harboring the D770_N771insG Exon20ins mutation (Supplementary Table S2), and YU-1163–bearing BALB/c nude mice models treated with amivantamab at 10 and 30 mg/kg dose, respectively. mF4/80 and mNKp46, markers of macrophages and NK cells in BALB/c nude mice, respectively, were elevated in tumors following treatment with amivantamab, suggesting that the mechanistic components of ADCC observed in vitro may translate to recruitment of key effector cells in tumors in vivo

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 9: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1202 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

(Fig. 5G; Supplementary Fig. S5B). In addition, these results suggest that amivantamab has greater ADCC and antitumor activity than cetuximab in the context of EGFR Exon20ins and that ADCC is an important mechanism in mediating the cytotoxic effects of amivantamab.

Amivantamab Demonstrates Antitumor Activity in a PDX Model Harboring the D770_N771insG Exon20ins Mutation

Treatment with amivantamab in the YHIM-1029 PDX model with D770_N771insG (Fig.  6A) resulted in a robust decrease in tumor volume, indicating that the antitumor activity observed in Ba/F3 and PDC models was preserved in a PDX model (Fig. 6B). In contrast, treatment with cetuximab (10 mg/kg) or poziotinib (1 mg/kg) only modestly reduced tumor volume. The dose of poziotinib was reduced to 1 mg/kg for this experiment due to the toxicity of poziotinib described above (Supplementary Fig.  S4D–S4H). Pharma-codynamic analysis showed that amivantamab treatment resulted in EGFR and MET downmodulation, inhibition of the downstream signaling pathways pAKT, pERK, and pS6, and increased markers of apoptosis (Fig.  6C). In contrast, tumors from mice treated with cetuximab or poziotinib main-tained the EGFR downstream signaling components pERK and pS6 (Fig.  6D), which was consistent with the modest

effects observed on tumor growth. Histopathologic examina-tion of tumor sections obtained following amivantamab or vehicle treatment using hematoxylin and eosin (H&E) staining, and IHC staining for EGFR, MET, and Ki-67, and terminal deoxynucleotidyl transferase–mediated dUTP nick end labe-ling (TUNEL) staining, further confirmed receptor inhibition and engagement of apoptotic machinery in EGFR Exon20ins–driven tumors in vivo (Fig. 6E). To verify whether the antitumor effect of amivantamab was affected by innate immunity in the in vivo models, we blocked the mouse CD16/CD32 via admin-istration of anti-CD16/CD32 antibodies. The antitumor effect of amivantamab shown in Fig.  6B was abrogated when the amivantamab-treated PDX-bearing BALB/c nude mice were cotreated with anti-CD16/CD32 antibodies, indicating that the antitumor effects of amivantamab were partially mediated by immune cells in this condition (Supplementary Fig. S6).

Antitumor Activity of Amivantamab in Patients with EGFR Exon20ins Disease

In an ongoing first-in-human study of amivantamab in patients with advanced NSCLC (NCT02609776), promis-ing clinical activity has been observed in patients with EGFR Exon20ins disease (13). A 58-year-old patient harboring the EGFR H773delinsNPY Exon20ins mutation achieved a partial response with a 65% tumor reduction (Fig. 7A), and a 48-year-old

D E H

F G I

0

1,100

1,000

900

800

700

600

500

400

300

200

100

0

100

200

300

400

500

600

700

800

900

0

0

100

−100

200

300

400

500

100200300400500600700800900

1,0001,1001,200

Vehicle Amivantamab30 mg/kg

Vehicle

Amivantamab30 mg/kg

pEGFR

pMET

MET

GAPDH

EGFR

Vehicle

#1 #2 #2#1

Amivantamab(30 mg/kg)

DFCI-127(P772_H773insPNP)

pEGFR

pMET

MET

GAPDH

EGFR

Vehicle

#1 #2 #2#1

Amivantamab(30 mg/kg)

YU-1163(S768_D770dup)

DFCI-127

YU-1163

DFCI-127(P772_H773insPNP)

YU-1163(S768_D770dup)

Tum

or v

olum

e (m

m3 )

Tum

or v

olum

e (m

m3 )

% c

hang

e in

tum

or v

olum

e%

cha

nge

in tu

mor

vol

ume

Vehicle

Amivantamab 30 mg/kg

Vehicle

Amivantamab 30 mg/kg

*

*

1

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

2 3

Day after treatment

Day after treatment

4 5 6 7 8 9 10 11

Figure 4. (Continued) Mice were treated with vehicle, IgG1 control, or amivantamab twice per week with intraperitoneal injections dosing 30 mg/kg. Data represent the mean ± SEM (n = 5/group). *, P < 0.0001 versus vehicle or IgG1 control. B, E, and G, A waterfall plot representation of the response of each tumor taken on the last day of treatment in the xenograft mice. C, H, and I, Tumor lysates of vehicle- or amivantamab-treated Ba/F3 cells or PDC xenograft mice were harvested and subjected to immunoblotting for pEGFR (Y1068), EGFR, pMET, and MET.

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 10: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1203

Figure 5.  Amivantamab has superior ADCC activity compared with cetuximab. A, Amivantamab-mediated ADCC activity against NSCLC PDCs express-ing EGFR Exon20ins mutations using PBMC (E:T ratio = 50: 1). ADCC assays were performed using DFCI-127 and YU-1163 PDCs as targets in the presence of IgG1 control, amivantamab, or cetuximab at various concentrations. PBMCs were cocultured for 4 hours with PDCs. *, P < 0.0001. Data are presented as averages ± SD of triplicate independent experiments. B, Amivantamab-mediated cytotoxicity against DFCI-127 and YU-1163 PDCs. PDCs were treated with IgG1, amivantamab (10 μg/mL), or cetuximab (10 μg/mL) for 24 hours in the presence or absence of PBMC (E:T ratio = 5:1). C, Quantitative analysis of the cells shown in the representative images (n = 3). *, P < 0.0001; **, P < 0.001. D, Pretreatment with FcR blocker with PBMC (E:T ratio = 50:1) reduced the amivantamab (10 μg/mL)-mediated ADCC effects. *, P < 0.0001. Data are presented as averages ± SD of triplicate independent experiments. E, IFNγ (pg/mL) levels in the cell culture media were detected by ELISA. The PDCs were cocultured with PBMCs in the presence of IgG1, amivantamab, or cetuximab at 1 μg/mL for 4 hours, and the culture medium was used for detection of IFNγ, *, P < 0.0001 versus cetuximab at the same concentration. F, PBMCs pretreated with FcR blocker reduced the IFNγ level in the culture medium in the presence of amivantamab (10 μg/mL), *, P < 0.0001; **, P < 0.001. Data are presented as averages ± SD of triplicate independent experiments. G, IHC staining for mF4/80 (macrophages) and mNKp46 (NK cells) of tumor sections in YHIM-1029 PDX-bearing BALB/c nude mice following 10 mg/kg IgG1 or 10 mg/kg amivantamab treatment.

A

00 0.01 0.1 1 10 100

Cyt

otox

icity

(%

)

Concentration (µg/mL)

Cetuximab(10 µg/mL)

Amivantamab(10 µg/mL)

IgG1(10 µg/mL)

DFCI-127

DFCI-127

− +

YU-1163

YU-1163

DFCI-127 YU-1163

Untre

ated

IgG1

Amiva

ntam

ab

IgG1

Amiva

ntam

ab

Cetux

imab

Cetux

imab

*

* *** **

* *

PBMC(E:T ratio = 5:1)

− +

0 0.01 0.1 1 10 100DFCI-127

IgG1

Amivantamab

Amivantamab +FcR blocker

**

*

*

YU-1163Concentration (µg/mL)

DFCI-127 YU-1163

IgG1 controlAmivantamabCetuximab

5

10

15

20

25

30

35

40

0

O.D

. (%

)Y

HIM

-102

9(D

770_

N77

1ins

G)

# 2

# 1

mF4/80

IgG1(10 mg/kg)

Amivantamab(10 mg/kg)

20 × 20 ×

# 2

# 1

20

40

60

80

100

120

140

IgG1

Untre

ated

IgG1

IgG1

Amiva

ntam

ab

Cextu

ximab

Untre

ated

Amiva

ntam

ab

Cextu

ximab

Amiva

ntam

ab

Cetux

imab

0

20

40

60

80

100

120

0

50

100

150

200

250

350

300

0

200

400

600

800

1,200

1,000

0

Cyt

otox

icity

(%

)

10

20

30

40

50

60

70

0

Cyt

otox

icity

(%

)

020406080

100120140160180

DFCI-127 YU-1163

Untre

ated

IgG1

Amiva

ntam

ab

Cetux

imab

*

0

200

PBMC −

PBMC +

− Fc blocker+ Fc blocker

400

600

800

1,000

1,200

1,400

IFN

γ (p

g/m

L)IF

(pg/

mL)

10

20

30

40

50

60

D

B E

C

G

F

mNKp46

IgG1(10 mg/kg)

Amivantamab(10 mg/kg)

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 11: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1204 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

Figure 6.  Amivantamab reduces tumors in a PDX model with D770_N771insG EGFR mutation. A, Sanger sequencing data depicting the D770_N771 insG mutations of the EGFR gene in a PDX model. B, Patient-derived tumors implanted in BALB/c nude mice were treated with vehicle, amivantamab (10 mg/kg), cetuximab (10 mg/kg), twice per week, intraperitoneal injections, or poziotinib (1 mg/kg), once a day. Data represent the mean ± SEM (n = 7/ group). *, P < 0.0001. Western blot analysis for the downstream signaling pathways of EGFR, MET, and apoptosis markers in tumors obtained from YHIM-1029 PDX models treated with (C) 10 mg/kg amivantamab, (D) 10 mg/kg cetuximab, or 1 mg/kg poziotinib. E, Histopathologic examination of tumor sections obtained from the PDX models following 10 mg/kg amivantamab or vehicle treatment. H&E staining and IHC staining for EGFR, pEGFR, MET, pMET, Ki-67, and TUNEL.

A YHIM-1029 (D770_N771insG) YHIM-1029(D770_N771insG)

YHIM-1029

TU

NE

LK

i-67

H&

EpM

ET

ME

TpE

GF

RE

GF

R

Vehicle

#1 #2 #1 #2

Amivantamab(10 mg/kg)

Amivantamab(10 mg/kg)

D770_N771insG

YHIM-1029

Day after treatment

Vehicle

*

**

Amivantamab 10 mg/kgCetuximab 10 mg/kgPoziotinib 1 mg/kg

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31

GACAAC GACGGGAAC

0

GAPDH

BIM

Cleaved PARP

Cleaved caspase-3

p53

p27

p21

pS6

ERK

pERK

pAKT

MET

pMET

pEGFR

EGFR

AKT

GAPDH

BIM

pS6

S6

ERK

pERK

pAKT

MET

pMET

pEGFR

EGFR

AKT

S6

Tum

or v

olum

e (m

m3 )

100200300400500600700800900

1,0001,1001,2001,3001,4001,500

C

B

D

E

YHIM-1029(D770_N771insG)

Vehicle

Vehicle

Cetuximab (10 mg/kg) Poziotinib (1 mg/kg)

#1 #2 #1 #2 #1 #2

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 12: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1205

Figure 7.  Amivantamab reduces tumors in patients with NSCLC with EGFR Exon20ins mutations. Radiologic response following amivantamab 1,050 mg treat-ment in (A) a 58-year-old patient with the EGFR H773delinsNPY mutation and (B) a 48-year-old patient with the EGFR S768_D770dup mutation. C, A proposed model of diverse antitumor mechanisms of amivantamab in NSCLC with EGFR Exon20ins.

CASE #1Pre PostAmivantamab

EGFR H773delinsNPYORR PR (−65% tumor reduction)

CASE #2Pre PostAmivantamab

NK cells

EGFRExon20ins

EGFRExon20ins

Osimertinib Gefitinib

O

O

O

NN

NNH

O

N

NNOH

N

N

N

Cl

P

PP

P

P

P P P

P

P

PP

P

F

HN

NSCLC with EGFR Exon20ins

MET

Amivantamab

Macrophage

Apoptosis

Direct inhibition oftumor growth mediatedby ADCC and ADCP

2

EGFR S768_D770dupORR PR (−38.9% tumor reduction)

B

A

C

Blockade of EGFR and METdownstream signaling pathway byinternalization of EGFR and MET

1

patient with the EGFR S768_D770dup Exon20ins mutation achieved a partial response with a 38.9% tumor reduction (Fig. 7B). These patients were progression-free for 92 and 32 weeks, respectively, on amivantamab, with manageable toxicities.

DiscussionIn our study, we characterized the antitumor activity of

amivantamab, a novel EGFR–MET bispecific antibody, in multiple preclinical models harboring EGFR Exon20ins

muta tions. In several Ba/F3 and PDC models expressing diverse EGFR Exon20ins mutations, amivantamab treatment resulted in EGFR and MET internalization, inhibition of downstream signaling cascades, engagement of apoptotic machinery, and subsequent inhibition of tumor cell prolif-eration. Importantly, these diverse action mechanisms of amivantamab were preserved in vivo as evidenced by phar-macodynamic analyses of tumors from cell line xenografts and PDX models treated with amivantamab (Fig.  7C). Fur-thermore, to the best of our knowledge, we first presented

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 13: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1206 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

evidence of clinical activity of amivantamab in two case stud-ies of patients with EGFR Exon20ins NSCLC from an ongo-ing phase I trial, highlighting the important translational nature of our preclinical work.

Currently, there are no targeted therapies approved for EGFR Exon20ins–positive advanced NSCLC. Owing to its small size and flexibility, poziotinib, an oral pan-HER inhibi-tor, has demonstrated greater activity than approved EGFR TKIs in vitro and in PDX models of EGFR Exon20ins–mutant NSCLC (18). In a single-center phase II trial, poziotinib showed a 43% confirmed RR in heavily pretreated advanced EGFR Exon20ins–mutant NSCLC (21). However, in a subse-quent pivotal phase II trial (NCT03318939), poziotinib yielded only 14.8% RR in a similar patient population (https://www.precisiononcologynews.com/drug-discovery-development/spectrums-poziotinib-failed-meet-primary-phase-ii-trial- endpoint#.XjJWkWgzaUk). TAK-788 has also shown preclinical activity against activating EGFR and HER2 mutations includ-ing EGFR Exon20ins. Although preliminary, in a phase I/II study (NCT02716116), TAK-788 produced clinical activity in a small subset of EGFR Exon20ins–mutant NSCLC (11). Importantly, treatment with poziotinib or TAK-788 was asso-ciated with a high incidence of EGFR WT–driven toxicity such as diarrhea and rashes, further limiting their clinical utility. Therefore, there is a substantial clinical need to identify new therapies for patients with EGFR Exon20ins.

In our study, amivantamab was clearly superior to pozi-otinib or cetuximab in terms of efficacy and tolerability in xenografts. Multiple facets of the MOA of amivantamab may contribute to the superior antitumor activity of amivantamab in the context of Exon20ins. In addition to the different MOA described in the various Exon20ins models described above, the ability of amivantamab to simultaneously bind two distinct epitopes of EGFR and MET may result in the concurrent interference with highly interconnected signaling pathways. To this end, amivantamab has been shown to have higher efficacy in decreasing tumor growth in the H1975-HGF model compared with the combination of anti-EGFR and anti-MET monovalent antibodies (36). This finding can be partly explained by an “avidity effect” whereby tumor cells expressing both targets, that is, EGFR and MET, bind to both arms of amivantamab with higher affinity than do cells that express only one target or engage a single Fab arm. A previous study has described correlations between binding affinity, receptor density, and receptor phosphorylation with amivantamab (37). Overall, bispecific antibodies show greater antitumor efficacy compared with a combination of monospe-cific mAbs via potential synergistic effects, and they increase selectivity by simultaneous targeting both receptors, favoring overexpressing cells as a consequence of avidity effects (38).

Amivantamab, produced by an engineered cell line defec-tive for protein fucosylation, has a low-level core fucosylation. The human FcγRIIIa, critical for ADCC, binds antibodies with low core fucosylation with higher affinity and consequently mediates more potent and effective NK cell–mediated killing of cancer cells (12). In this study, amivantamab demonstrated more robust ADCC in EGFR Exon20ins mutation models than cetuximab, an EGFR-directed antibody that has not shown robust utility in NSCLC (39, 40). This ADCC activity was correlated with secreted IFNγ levels. It is possible that the

IFNγ secreted from amivantamab mediated active immune cells, including NK cells and macrophages, may restimulate and recruit surrounding immune cells to the tumor, although additional studies are required to investigate this hypothesis. In addition, immunocytokines secreted from NK cells lead to upregulation of ICAM1 on target cells, rendering them more susceptible to target cell cytolysis (41). Indeed, phar-macodynamic analyses of tumors from mice treated with amivantamab revealed increased tumor levels of NK cells and macrophages. In contrast to many other therapeutic antibod-ies used in the clinical setting, amivantamab was designed and engineered with a low fucose backbone, which enhances its binding to FcγRIIIa (17), which is present on NK cells, mono-cytes, and macrophages. The human FcγRIIIa, critical for ADCC, binds antibodies with low-level core fucosylation more tightly and consequently mediates more potent and effective ADCC killing of cancer cells (42). Thus, the enhanced binding of amivantamab to FcγRIIIa may lead to increased induction of Fc effector functions in comparison with other (normal fucose) hIgG1 antibodies such as cetuximab.

In this study, we used two different xenograft models, NOG mice and BALB/c nude mice for in vivo study. As demonstrated in many studies, NOG mice have impaired innate immunity and extremely low NK-cell activity, whereas BALB/c nude mice have intact innate immunity and active NK cells (43, 44). For this reason, more potent ADCC activ-ity was expected in BALB/c nude mice than NOG mice. Therefore, a minimal amivantamab-mediated ADCC activity resulted in the modest efficacy with amivantamab in Ba/F3- and DFCI-127–bearing NOG mice (Fig.  4A and D; Supple-mentary Fig. S4A). On the other hand, the significant tumor regression was observed with amivantamab in YU-1163–bear-ing and YHIM-1029–bearing BALB/c nude mice (Figs. 4F and 6B), which resulted from multiple MOA of amivantamab to block the EGFR and MET downstream signaling pathway and elicit ADCC.

It has been observed that EGFR and tumor suppressor TP53 genes are commonly mutated in patients with NSCLC with independent prognostic implications. Furthermore, in patients with concomitant mutations in EGFR and TP53, there have been reports of decreased responsiveness to EGFR TKIs (45). A similar effect was observed in our study in the YU-1163 PDC and YUO-029 PDO following treatment with amivantamab. On the other hand, amivantamab exhibited a potent in vivo activity in YU-1163–bearing BALB/c nude mice, suggesting that ADCC activity of amivantamab, shown in Fig. 5A–G, was believed to be involved in the in vivo antitumor activity. These results suggest that the combination of effector cell–dependent and -independent MOAs elicited by amivan-tamab (Fig.  7C) may result in antitumor activity in tumors harboring a coalescence of intractable mutations, for exam-ple in EGFR Exon20ins disease, and concomitant deleterious mutations such as the TP53 mutation present in our preclini-cal model and reported in the broader patient population.

In our study, amivantamab demonstrated less skin tox-icity than poziotinib. Amivantamab-treated BALB/c nude mice appeared phenotypically normal with only minimal signs of keratosis on the face, indicating that amivantamab was well tolerated in this preclinical model. In contrast, treatment with poziotinib resulted in severe keratosis,

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 14: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1207

significant weight loss, and even sudden death (Supplemen-tary Fig. S4D–S4H).

In conclusion, our data demonstrated that amivantamab functions through multiple MOA to elicit antitumor activity in multiple preclinical models of EGFR Exon20ins disease. Consequently, amivantamab warrants further clinical inves-tigation, as evidenced by clinical results from 2 patients with EGFR Exon20ins NSCLC who have been treated with ami-vantamab in the clinical setting. This represents important progress toward the identification of an effective therapeutic option for patients with NSCLC with EGFR Exon20ins, an area of high unmet medical need.

MethoDsBa/F3 Cell Lines and Drug Compounds

All mutant Ba/F3 cell lines were purchased from the German Collec-tion of Microorganisms and Cell Cultures and were obtained from the Dana-Farber Cancer Institute, Harvard University (Boston, MA). All cells were maintained in RPMI1640 medium supplemented with 10% FBS and puromycin in a humidified incubator with 5% CO2. Amivan-tamab and IgG1 controls were provided by Janssen. Gefitinib, osimer-tinib, cetuximab, and poziotinib were purchased from SelleckChem.

AntibodiesPrimary antibodies specific for pEGFR (2234), EGFR (4267), pMET

(3077), MET (8198), pERK (4370), ERK (9107), pAKT (9271), AKT (9272), pS6 (4858), S6 (2217), p27 (2252), cleaved PARP (5625S), cleaved caspase-3 (9661), and BIM (2933) were purchased from Cell Signaling Technology; p21(sc-817) and p53 (sc-126) were purchased from Santa Cruz Biotechnology, Inc.; and GAPDH (PAB13195) was purchased from Abnova. For the IHC assay, mF4/80 (#70076) and mNKp46 (AF2225) were purchased from Cell Signaling Technology and R&D Systems, respectively.

PDCsYU-1163 (S768_D770dup) cell lines were derived from malignant

effusions from patients with NSCLC and cultured on collagen-coated plates in ACL-4 medium supplemented with 5% FBS. The cells main-tained the driver oncogenes that were observed in the patients. Cells were enriched in an epithelial cell adhesion molecule (EPCAM)– positive cell population with a purity of over 95% before they were sub-jected to further assays. DFCI-58 (H773_V774insNPH) and DFCI-127 (P772_H773insPNP) cell lines were obtained from the Dana-Farber Cancer Institute and were cultured in ACL-4 medium and RPMI medium with 10% FBS, respectively. All patient samples were collected after written informed consent from the patients was obtained. The study protocols were approved by the respective institutional review boards.

PDO CulturePDOs (YUO-029 and YUO-036) were established as described pre-

viously (46). Briefly, malignant effusions from 2 patients with NSCLC were collected and centrifuged, and the cell pellets were mixed with growth factor–reduced Matrigel (Corning) and seeded into 48-well plates. Solidified gels were overlaid with advanced DMEM/F12 (Inv-itrogen) containing 1× Glutamax (Invitrogen), 10 mmol/L HEPES (Invitrogen), 1× Antibiotic-Antimycotic (Invitrogen), 1× B-27 (Inv-itrogen), 20% R-spondin conditioned medium, 5 mmol/L Nicotina-mide (Sigma), 1.25 mmol/L N-acetylcysteine (Sigma), 500 nmol/L SB-202190 (Sigma), 500 nmol/L A83-01 (Tocris), 100 ng/mL Mouse Noggin (PeproTech), 100 ng/mL human FGF10 (PeproTech), 25 ng/mL human FGF7 (PeproTech), 50 μg/mL Primocin (InvivoGen), and

10 μmol/L Y-27632 (Enzo). R-spondin conditioned medium was produced from HA-R-Spondin1-Fc 293T Cells (Amsbio). For passag-ing, organoids were collected, mechanically sheared with a 25-gauge needle, and washed with cold PBS before the organoid pellets were resuspended in the Matrigel and seeded into 24-well plates at ratios of 1:2 to 1:4. The culture medium was replenished at least twice a week. Cell viability tests were performed as described previously (47). Briefly, organoids were trypsinized into single cells and cultured for 5 to 10 days. Then, the organoids were collected, resuspended in the medium containing 5% Matrigel, and plated in a 96-well plate (Corning) at a concentration of 2,000 organoids/μL. The medium with the IgG1 control or amivantamab at diverse concentrations was added and incubated for 72 hours. Cell viability was measured using CellTiter-Glo 3D Culture Reagent (Promega) on a microplate lumi-nometer according to the manufacturer’s instructions.

PDX ModelsPDXs were created using 6- to 8-week-old female SCID (NOG) and

nude (nu/nu) mice obtained from Orient Bio. All methods complied with the guidelines of our Institutional Animal Research Committee (Yonsei University College of Medicine, Seoul, Republic of South Korea) and were approved by the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). After removal of the necrotic and supporting tissues from core biopsy specimens, small specimens of the tumor tissue (3 mm × 3 mm × 3 mm) from each patient were implanted subcutaneously in 1 to 2 mice. After the tumor reached 1.5 cm in diameter, it was excised, dissected into small specimens (3 mm × 3 mm × 3 mm), and reimplanted into nude mice.

In Vivo Xenograft StudiesFemale athymic BALB-c/nu mice were obtained from Orient Bio

at 5 to 6 weeks of age. All mice were handled in accordance with the Animal Research Committee’s Guidelines at Yonsei University Col-lege of Medicine, and all facilities were approved by AAALAC. Ba/F3 cells and PDCs (1 × 107 cells) were injected subcutaneously into the NOG and BALB-c/nu mice, respectively, and growth was measured twice weekly; after establishment of palpable lesions, mice were assigned to testing. Once the tumor volume reached approximately 150 to 200 mm3, mice were randomly allocated into groups of 5 animals to receive either vehicle, IgG1 control, or amivantamab. The tumor size was measured every 2 days using calipers. The average tumor volume in each group was expressed in mm3 and calculated according to the equation for a prolate spheroid: tumor volume = 0.523 × (large diameter) × (small diameter)2.

Antiproliferation AssayBa/F3 cells or PDCs expressing EGFR Exon20ins mutations were

seeded onto 96-well plates in 100 μL. After treatment with IgG1 control, amivantamab, gefitinib, or osimertinib for 72 hours, cell viability was measured by quantifying the total amount of ATP using the CellTiter-Glo 2.0 Assay Kit (Promega) according to the manufac-turer’s instructions.

Colony Formation AssayCells were seeded onto 6-well culture plates and incubated for

12 days at 37°C with amivantamab (0, 0.1, or 1 mg/mL). Cells were washed with PBS, fixed, and stained with 4% paraformaldehyde in 5% crystal violet for 10 minutes. Colonies were eluted with 1% SDS, and the optical density value was determined using ELISA at 470 nm.

ADCC AssaysThe ADCC assay was conducted using the Lactase Dehydrogenase

(LDH) Cytotoxicity Detection Kit (Roche) in accordance with the manufacturer’s instructions. Human PBMCs obtained from healthy

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 15: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Yun et al.RESEARCH ARTICLE

1208 | CANCER DISCOVERY AUGUST 2020 AACRJournals.org

volunteers were used as the effector cells. ADCC was conducted using an E:T cell ratio ranging from 50:1 to 5:1 and incubated for 4 to 24 hours at 37°C in 5% CO2. Amivantamab concentrations of 100 to 0.01 μg/mL were tested. The LDH activity of the cell culture supernatants was measured, and the percentage cytotoxicity was calculated as described in the manufacturer’s protocol.

IF AnalysisPDCs were seeded on 0.01% poly-L-lysine (Sigma-Aldrich) coated

coverslips. The following day, cells were treated with IgG1 control or amivantamab at 0.1 mg/mL. After 72 hours, the coverslips were fixed in 4% formaldehyde for 15 minutes, permeabilized with 0.5% Triton X-100 for 5 minutes, and incubated with primary antibody for 1 hour at room temperature. The primary antibodies used in the study were rabbit monoclonal anti-EGFR and anti-MET (Santa Cruz Biotechnology) and ab992 (Millipore) at a dilution of 1:100. The cov-erslips were rinsed twice with PBS, followed by incubation with the appropriate fluorophore-conjugated secondary antibody (Invitrogen) for 1 hour at room temperature. The cells were counterstained with 4′,6-diamidino-2-phenylindole (DAPI; 300 nmol/L; Invitrogen), and the coverslips were mounted on slides using Faramount Aqueous Mounting Medium (Dako).

IHCIHC was performed using the Automated Staining System (BOND

Rx, Leica Biosystems). Briefly, 4-mm paraffin-embedded tumor sections were deparaffinized and rehydrated. Slides then under-went heat-induced epitope retrieval with citrate buffer at 100°C for 20 minutes. Antibodies were used at 1:100 dilution and hematoxylin solution was used for counterstaining. Stained slides were visualized with a Vectra Polaris and the Phenochart program.

In Vivo Pharmacodynamic StudyMice bearing tumor tissues were treated with vehicle, IgG1 control,

or amivantamab (10 or 30 mg/kg) twice per week intraperitoneally, or cetuximab (10 mg/kg) or poziotinib (1 mg/kg) once daily. The tumor samples were collected 48 hours after 15 days of treatment, and EGFR and MET downstream signaling was evaluated by immunoblotting.

PatientsThe clinical study (NCT02609776) was approved by the appro-

priate institutional review boards at each participating site and conducted in accordance with Good Clinical Practice guidelines and the ethical principles of the Declaration of Helsinki. All patients pro-vided written informed consent.

Statistical AnalysisData were collected from three independent experiments and

either presented descriptively or analyzed by one-way ANOVA, fol-lowed by the Dunnett test or Student t test. Dose–response curves were prepared using GraphPad Prism (Ver. 5, GraphPad Software Inc.).

Disclosure of Potential Conflicts of InterestJ.C. Curtin is an associate director, oncology translational research

at Janssen Research and Development. R.E. Knoblauch is an executive medical director at Janssen. M.V. Lorenzi is vice president, oncology at Janssen, Pharmaceutical Companies of Johnson and Johnson and has ownership interest (including patents) in Johnson & Johnson. B.C. Cho is a professor at Novartis, AstraZeneca, Takeda, Janssen, Medpacto, Blueprint Medicines, MSD, Boehringer-Ingelheim, Roche, BMS, Ono, Yuhan, Pfizer, Eli Lilly, and Janssen, reports receiving commercial research grants from Novartis, Bayer, MSD, AbbVie, Medpacto, GIInnovation, Eli Lilly, Blueprint Medicines, AstraZeneca,

MOGAM Institute, Dong-A ST, Champions Oncology, Janssen, Yuhan, Ono, and Dizal Pharma, reports receiving speakers bureau honoraria from Novartis, Bayer, MSD, AstraZeneca, MOGAM Insti-tute, Dong-A ST, Champions Oncology, Janssen, Yuhan, Ono, and Dizal Pharma, has ownership interest (including patents) in Thera-CanVac Inc, Gencurix Inc, Bridgebio Therapeutics, and KANAPH Therapeutic Inc, and has received other remuneration from Daan Biotherapeutics. No potential conflicts of interest were disclosed by the other authors.

Authors’ ContributionsConception and design: J. Yun, J.-H. Kim, S.M. Lim, M.V. Lorenzi, B.C. ChoDevelopment of methodology: J. Yun, J.-H. Kim, S.M. Lim, B.C. ChoAcquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J. Yun, S.-Y. Kim, J.-H. Kim, M.H. Hong, H.R. Kim, R.E. Knoblauch, B.C. ChoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J. Yun, J.-H. Kim, K.-H. Pyo, S.G. Heo, S. Lim, J.C. Curtin, R.E. Knoblauch, B.C. ChoWriting, review, and/or revision of the manuscript: J. Yun, S.-H. Lee, M.R. Yun, M. Thayu, J.C. Curtin, R.E. Knoblauch, M.V. Lorenzi, A. Roshak, B.C. ChoAdministrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.-H. Lee, S.-Y. Jeong, J.-H. Kim, C.-W. Park, B.C. ChoStudy supervision: S.M. Lim, B.C. Cho

AcknowledgmentsThis research was supported by the Basic Science Research Pro-

gram through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (NRF-2016R1A2B3016282 and 2018R1C1B6008916), Republic of South Korea.

Received January 31, 2020; revised April 17, 2020; accepted May 7, 2020; published first May 15, 2020.

REFERENCES 1. Fang WF, Huang YH, Hong SD, Zhang ZH, Wang MH, Gan JD, et al.

EGFR exon 20 insertion mutations and response to osimertinib in non-small-cell lung cancer. BMC Cancer 2019;19:595.

2. Nagano T, Tachihara M, Nishimura Y. Mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitors and a potential treatment strategy. Cells 2018;7:212.

3. Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014;311:1998–2006.

4. Travis WD. 2015 WHO classification of the pathology and genetics of tumors of the lung. J Thorac Oncol 2015;10:S68.

5. Gazdar AF. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 2009;28:S24–S31.

6. Vyse S, Huang PH. Targeting EGFR exon 20 insertion mutations in non-small cell lung cancer. Signal Transduct Target Ther 2019;4:5.

7. Yasuda H, Park E, Yun CH, Sng NJ, Lucena-Araujo AR, Yeo WL, et al. Structural, biochemical, and clinical characterization of epidermal growth factor receptor (EGFR) exon 20 insertion mutations in lung cancer. Sci Transl Med 2013;5:216ra177.

8. Riess JW, Gandara DR, Frampton GM, Madison R, Peled N, Bufill JA, et  al. Diverse EGFR exon 20 insertions and co-occurring molecular alterations identified by comprehensive genomic profiling of NSCLC. J Thorac Oncol 2018;13:1560–8.

9. Oxnard GR, Lo PC, Nishino M, Dahlberg SE, Lindeman NI, Butaney M, et al. Natural history and molecular characteristics of

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 16: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

Amivantamab in NSCLC with EGFR Exon20ins RESEARCH ARTICLE

AUGUST 2020 CANCER DISCOVERY | 1209

lung cancers harboring EGFR exon 20 insertions. J Thorac Oncol 2013;8:179–84.

10. Yang ZD, Tchekmedyian N, Chu DT, Reddy G, Bhat G, Socinski MA. A phase 2 study of poziotinib in patients with EGFR or HER2 exon 20 mutation-positive non-small cell lung cancer. J Clin Oncol 36:15s, 2018 (suppl; abstr TPS9106).

11. Janne PA, Neal JW, Camidge DR, Spira AI, Piotrowska Z, Horn L, et al. Antitumor activity of TAK-788 in NSCLC with EGFR exon 20 inser-tions. J Clin Oncol 2019;37:9007.

12. Cho BC, Lee JS, Han JY, Cho EK, Haura E, Lee KH, et al. JNJ-61186372 (JNJ-372), an EGFR-cMET bispecific antibody, in advanced non-small cell lung cancer (NSCLC): an update on phase I results. Ann Oncol 2018;29:viii493–547.

13. Haura EB, Cho BC, Lee JS, Han JY, Lee KH, Sanborn RE, et al. JNJ-61186372 (JNJ-372), an EGFR-cMet bispecific antibody, in EGFR-driven advanced non-small cell lung cancer (NSCLC). J Clin Oncol 37:15s, 2019 (suppl; abstr 9009).

14. Moores SL, Chiu ML, Bushey BS, Chevalier K, Luistro L, Dorn K, et al. A novel bispecific antibody targeting EGFR and cMet is effective against EGFR inhibitor-resistant lung tumors. Cancer Res 2016;76: 3942–53.

15. Kosaka T, Tanizaki J, Paranal RM, Endoh H, Lydon C, Capelletti M, et al. Response heterogeneity of EGFR and HER2 exon 20 insertions to covalent EGFR and HER2 inhibitors. Cancer Res 2017;77:2712–21.

16. Frega S, Lorenzi M, Fassan M, Indraccolo S, Calabrese F, Favaretto A, et al. Clinical features and treatment outcome of non-small cell lung cancer (NSCLC) patients with uncommon or complex epidermal growth factor receptor (EGFR) mutations. Oncotarget 2017;8:32626–38.

17. Grugan KD, Dorn K, Jarantow SW, Bushey BS, Pardinas JR, Laquerre S, et al. Fc-mediated activity of EGFR x c-Met bispecific antibody JNJ-61186372 enhanced killing of lung cancer cells. Mabs 2017;9:114–26.

18. Robichaux JP, Elamin YY, Tan Z, Carter BW, Zhang SX, Liu SW, et al. Mechanisms and clinical activity of an EGFR and HER2 exon 20-selective kinase inhibitor in non-small cell lung cancer. Nat Med 2018;24:638–46.

19. Colwell J. Poziotinib shows promise for rare lung cancer. Cancer Discov 2018;8:OF4.

20. Cha MY, Lee KO, Kim M, Song JY, Lee KH, Park J, et al. Antitumor activity of HM781-36B, a highly effective pan-HER inhibitor in erlotinib-resistant NSCLC and other EGFR-dependent cancer mod-els. Int J Cancer 2012;130:2445–54.

21. Heymach J, Negrao M, Robichaux J, Carter B, Patel A, Altan M, et al. A phase II trial of poziotinib in EGFR and HER2 exon 20 mutant non-small cell lung cancer (NSCLC). J Thorac Oncol 2018;13:S323–S4.

22. Costa DB, Halmos B, Kumar A, Schumer ST, Huberman MS, Boggon TJ, et  al. BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations. PLoS Med 2007;4:1669–80.

23. Shi PY, Oh YT, Deng L, Zhang GJ, Qian GQ, Zhang S, et al. Overcom-ing acquired resistance to AZD9291, a third-generation EGFR inhibi-tor, through modulation of MEK/ERK-dependent Bim and Mcl-1 degradation. Clin Cancer Res 2017;23:6567–79.

24. Canale M, Petracci E, Delmonte A, Chiadini E, Dazzi C, Papi M, et al. Impact of TP53 mutations on outcome in EGFR-mutated patients treated with first-line tyrosine kinase inhibitors. Clin Cancer Res 2017;23:2195–202.

25. Conde E, Angulo B, Tang MY, Morente M, Torres-Lanzas J, Lopez-Encuentra A, et  al. Molecular context of the EGFR mutations: evi-dence for the activation of mTOR/S6K signaling. Clin Cancer Res 2006;12:710–7.

26. Lin CL, Liang YJ, Zhu HL, Zhang JJ, Zhong XY. R280T mutation of p53 gene promotes proliferation of human glioma cells through GSK-3 beta/PTEN pathway. Neurosci Lett 2012;529:60–5.

27. Zhu HB, Yang K, Xie YQ, Lin YW, Mao QQ, Xie LP. Silencing of mutant p53 by siRNA induces cell cycle arrest and apoptosis in human bladder cancer cells. World J Surg Oncol 2013;11:22.

28. Mantovani F, Collavin L, Del Sal G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ 2019;26:199–212.

29. Alam SK, Yadav VK, Bajaj S, Datta A, Dutta SK, Bhattacharyya M, et  al. DNA damage-induced ephrin-B2 reverse signaling promotes chemoresistance and drives EMT in colorectal carcinoma harboring mutant p53. Cell Death Differ 2016;23:707–22.

30. Sauer L, Gitenay D, Vo C, Baron VT. Mutant p53 initiates a feedback loop that involves Egr-1/EGF receptor/ERK in prostate cancer cells. Oncogene 2010;29:2628–37.

31. Sunada H, Magun BE, Mendelsohn J, Macleod CL. Monoclonal-antibody against epidermal growth-factor receptor is internalized without stimulating receptor phosphorylation. Proc Natl Acad Sci U S A 1986;83:3825–9.

32. Okada Y, Kimura T, Nakagawa T, Okamoto K, Fukuya A, Goji T, et al. EGFR downregulation after anti-EGFR therapy predicts the antitu-mor effect in colorectal cancer. Mol Cancer Res 2017;15:1445–54.

33. Wang W, Erbe AK, Hank JA, Morris ZS, Sondel PM. NK cell-mediated antibody-dependent cellular cytotoxicity in cancer immunotherapy. Front Immunol 2015;6:368.

34. Fauriat C, Long EO, Ljunggren HG, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 2010;115:2167–76.

35. Reefman E, Kay JG, Wood SM, Offenhauser C, Brown DL, Roy S, et al. Cytokine secretion is distinct from secretion of cytotoxic granules in NK cells. J Immunol 2010;184:4852–62.

36. Zheng S, Moores S, Jarantow S, Pardinas J, Chiu M, Zhou H, et  al. Cross-arm binding efficiency of an EGFR X C-Met bispecific anti-body. Clin Pharmacol Ther 2017;101:S14.

37. Jarantow SW, Bushey BS, Pardinas JR, Boakye K, Lacy ER, Sanders R, et al. Impact of cell-surface antigen expression on target engagement and function of an epidermal growth factor receptor x c-MET bispe-cific antibody. J Biol Chem 2015;290:24689–704.

38. Sellmann C, Doerner A, Knuehl C, Rasche N, Sood V, Krah S, et al. Balancing selectivity and efficacy of bispecific epidermal growth fac-tor receptor (EGFR) x c-MET antibodies and antibody-drug conju-gates. J Biol Chem 2016;291:25106–19.

39. Wong SF. Cetuximab: an epidermal growth factor receptor monoclo-nal antibody for the treatment of colorectal cancer. Clin Ther 2005; 27:684–94.

40. Ramalingam S, Forster J, Naret C, Evans T, Sulecki M, Lu HL, et al. Dual inhibition of the epidermal growth factor receptor with cetuxi-mab, an IgG1 monoclonal antibody, and gefitinib, a tyrosine kinase inhibitor, in patients with refractory non-small cell lung cancer (NSCLC): a phase I study. J Thorac Oncol 2008;3:258–64.

41. Wang RP, Jaw JJ, Stutzman NC, Zou ZC, Sun PD. Natural killer cell-produced IFN-γ and TNF-α induce target cell cytolysis through up-regulation of ICAM-1. J Leukocyte Biol 2012;91:299–309.

42. Satoh M, Iida S, Shitara K. Non-fucosylated therapeutic antibod-ies as next-generation therapeutic antibodies. Expert Opin Biol Th 2006;6:1161–73.

43. Puchalapalli M, Zeng XK, Mu L, Anderson A, Glickman LH, Zhang M, et al. NSG mice provide a better spontaneous model of breast cancer metastasis than athymic (Nude) mice. PLoS One 2016;11:e0163521.

44. Okada S, Vaeteewoottacharn K, Kariya R. Application of highly immunocompromised mice for the establishment of patient-derived xenograft (PDX) models. Cells 2019;8:889.

45. Aggarwal C, Davis CW, Mick R, Thompson JC, Ahmed S, Jeffries S, et  al. Influence of TP53 mutation on survival in patients with advanced EGFR-mutant non-small-cell lung cancer. JCO Precis Oncol 2018;2018:10.1200/PO.18.00107.

46. Sachs N, Papaspyropoulos A, Zomer-van Ommen DD, Heo I, Bot-tinger L, Klay D, et al. Long-term expanding human airway organoids for disease modeling. EMBO J 2019;38:e100300.

47. Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 2018;556:457–62.

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116

Page 17: Antitumor Activity of Amivantamab (JNJ-61186372), an ......Jiyeon Yun1, Soo-Hwan Lee2, Seok-Young Kim1, Seo-Yoon Jeong1, Jae-Hwan Kim1, Kyoung-Ho Pyo1, Chae-Won Park 1 , Seong Gu Heo

2020;10:1194-1209. Published OnlineFirst May 15, 2020.Cancer Discov   Jiyeon Yun, Soo-Hwan Lee, Seok-Young Kim, et al.  

Driven NSCLC−20 Insertion Exon EGFRMET Bispecific Antibody, in Diverse Models of −

Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR

  Updated version

  10.1158/2159-8290.CD-20-0116doi:

Access the most recent version of this article at:

  Material

Supplementary

  1

http://cancerdiscovery.aacrjournals.org/content/suppl/2020/05/15/2159-8290.CD-20-0116.DCAccess the most recent supplemental material at:

   

   

  Cited articles

  http://cancerdiscovery.aacrjournals.org/content/10/8/1194.full#ref-list-1

This article cites 46 articles, 14 of which you can access for free at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  SubscriptionsReprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerdiscovery.aacrjournals.org/content/10/8/1194To request permission to re-use all or part of this article, use this link

Cancer Research. on January 24, 2021. © 2020 American Association forcancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst May 15, 2020; DOI: 10.1158/2159-8290.CD-20-0116