ArticlePDF AvailableLiterature Review

Amivantamab-Vmjw: A Novel Treatment for Patients with NSCLC Harboring EGFR Exon 20 Insertion Mutation after Progression on Platinum-Based Chemotherapy

MDPI
Biomedicines
Authors:

Abstract and Figures

Objective: This study is a comprehensive review of the clinical pharmacology, pharmacokinetics, efficacy, safety, and clinical applicability of amivantamab-vmjw for metastatic non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) exon 20 insertion (exon20ins) mutation. Data synthesis: The literature search to identify clinical trials returned only the CHRYSALIS phase 1 study. In a phase I trial, amivantamab-vmjw was associated with an overall response rate (ORR) of 40% (95% CI, 29-51) in the EGFR exon20ins NSCLC patient population (n = 81) after platinum-based chemotherapy. There were 3 complete responses (CRs) and 29 partial responses (PRs). The median duration of response (DOR) was 11.1 months (95% CI, 6.9-not reached; NR). The median progression-free survival (PFS) was 8.3 months (95% CI, 6.5-10.9), and overall survival (OS) was 22.8 months (95% CI, 14.6-NR). Application to clinical practice: This review summarizes the pharmacology, clinical evidence, and use of amivantamab-vmjw for patients with locally advanced or metastatic NSCLC with EGFR exon20ins mutation. Conclusion: The FDA approval of amivantamab-vmjw, the first bispecific antibody to target the exon20ins mutation, represents an important advancement in the treatment of patients with NSCLC with limited effective treatment options. The initial findings of the CHRYSALIS trial demonstrate an overall tumor response benefit with an acceptable safety profile.
Content may be subject to copyright.
Citation: Shah, V.; McNatty, A.;
Simpson, L.; Ofori, H.; Raheem, F.
Amivantamab-Vmjw: A Novel
Treatment for Patients with NSCLC
Harboring EGFR Exon 20 Insertion
Mutation after Progression on
Platinum-Based Chemotherapy.
Biomedicines 2023,11, 950.
https://doi.org/10.3390/
biomedicines11030950
Academic Editor: Bruno
Kaufmann Robbs
Received: 26 February 2023
Revised: 11 March 2023
Accepted: 15 March 2023
Published: 20 March 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
biomedicines
Review
Amivantamab-Vmjw: A Novel Treatment for Patients with
NSCLC Harboring EGFR Exon 20 Insertion Mutation after
Progression on Platinum-Based Chemotherapy
Vishal Shah 1,* , Andrea McNatty 2, Lacey Simpson 1, Henry Ofori 1and Farah Raheem 1
1Department of Pharmacy, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, USA
2Seagen, Bothell, WA 85054, USA
*Correspondence: shah.vishal@mayo.edu
Abstract:
Objective: This study is a comprehensive review of the clinical pharmacology, pharmacoki-
netics, efficacy, safety, and clinical applicability of amivantamab-vmjw for metastatic non-small cell
lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) exon 20 insertion (exon20ins) mu-
tation. Data Synthesis: The literature search to identify clinical trials returned only the CHRYSALIS
phase 1 study. In a phase I trial, amivantamab-vmjw was associated with an overall response rate
(ORR) of 40% (95% CI, 29–51) in the EGFR exon20ins NSCLC patient population (n = 81) after
platinum-based chemotherapy. There were 3 complete responses (CRs) and 29 partial responses
(PRs). The median duration of response (DOR) was 11.1 months (95% CI, 6.9—not reached; NR).
The median progression-free survival (PFS) was 8.3 months (95% CI, 6.5–10.9), and overall survival
(OS) was 22.8 months (95% CI, 14.6—NR). Application to Clinical Practice: This review summa-
rizes the pharmacology, clinical evidence, and use of amivantamab-vmjw for patients with locally
advanced or metastatic NSCLC with EGFR exon20ins mutation. Conclusion: The FDA approval
of amivantamab-vmjw, the first bispecific antibody to target the exon20ins mutation, represents an
important advancement in the treatment of patients with NSCLC with limited effective treatment
options. The initial findings of the CHRYSALIS trial demonstrate an overall tumor response benefit
with an acceptable safety profile.
Keywords: amivantamab-vmjw; lung cancer; EGFR; exon 20; NSCLC
1. Introduction
Lung cancer is one of the most common types of cancer among both males and fe-
males, accounting for 25% of all cancer-related deaths [
1
3
]. NSCLC accounts for 85% of all
diagnosed types of lung cancer [
1
,
4
]. NSCLC is biologically and molecularly diverse, and
targeted therapies against oncogenic driver mutations have become pivotal in improving
survival outcomes [
1
,
5
]. Due to the heterogeneous nature of NSCLC, over 600 EGFR muta-
tion variants have been described [
6
]. Driver mutations within the EGFR exons
18–21 occur
in up to 40% of NSCLC cases [
6
,
7
]. Point mutations in exons 18, 19, and 21 represent
85% of EGFR mutation-positive NSCLC cases and have been shown to be responsive to
targeted treatment using EGFR-specific tyrosine kinase inhibitors (TKIs) [
8
,
9
]. The next
most frequently occurring EGFR mutation is exon20ins, accounting for approximately 12%
of cases.
EGFR exon20ins mutations are defined by in-frame insertions and duplications near
the EGFR kinase domain. EGFR exon20ins mutations contain a modified active site that
hinders the binding of currently approved EGFR TKI molecules, resulting in low response
rates of <9%. [
8
,
9
] Due to this low response rate, the median OS in NSCLC with EGFR
exon20ins mutations is only 16 months [
10
14
]. The absence of tumor response to standard
EGFR TKIs and suboptimal efficacy of chemotherapy highlight the cruciality of identifying
targeted therapy with activity against EGFR exon20ins mutations in NSCLC.
Biomedicines 2023,11, 950. https://doi.org/10.3390/biomedicines11030950 https://www.mdpi.com/journal/biomedicines
Biomedicines 2023,11, 950 2 of 11
Amivantamab-vmjw presents a novel mechanism as a bispecific EGFR and mes-
enchymal epithelial transition (MET) receptor antibody with accelerated FDA approval
in May 2021 for patients with metastatic NSCLC with EGFR exon20ins mutations who
have progressed after platinum-based chemotherapy [
15
]. The accelerated approval was
based on the positive results of overall response rate as well as the duration of response
demonstrated by an open-label, phase I clinical trial [
8
]. In this article, we describe the
pharmacology, clinical efficacy, and safety of amivantamab-vmjw for treatment of EGFR
exon20ins mutation-positive NSCLC.
2. Pharmacology
Acquired resistance to standard of care treatments can develop in NSCLC due to
mutations in EGFR and activation of the MET pathway. EGFR and MET pathways are
required for tumor cell proliferation and growth. The mutations in the EGFR protein
activate a chain of events that constitutes subsequent growth and survival signaling in
cancer cells [
7
,
9
]. EGFR mutations develop in up to 85% of patients with NSCLC [
8
].
Driver mutations in the exons of the EGFR tyrosine kinase domain have been evolving
targets for NSCLC therapies. First-, second-, and third-generation EGFR TKIs have been
established as standard of care treatment for NSCLC with EGFR mutations except for
exon20ins mutations. EGFR exon20ins mutation is a distinct subtype of EGFR oncogenic
mutations that has been shown to be unresponsive to third-generation EGFR TKIs [
8
,
9
].
The use of EGFR TKIs can also induce MET receptor amplification, which bypasses the
EGFR pathway, causing signaling downstream to promote cancer cell proliferation and
resistance to standard of care treatments [16].
Amivantamab-vmjw demonstrates a novel mechanism of action as a fully humanized
immunoglobulin G1-based bispecific antibody that targets the domains of EGFR and MET.
In vivo
and
in vitro
studies of amivantamab-vmjw demonstrated a dose-dependent induc-
tion of EGFR and MET downregulation, antiproliferative effect, and antibody-dependent
cellular cytotoxicity [
15
]. Antitumor effects were shown with both Fc receptor-independent
and -dependent mechanisms. Fc-independent functions block EGFR and MET signal-
ing via ligand binding and receptor inactivation. Amivantamab-vmjw binding to the
exon20ins-mutated EGFR tyrosine kinase receptor results in degradation of EGFR and
MET receptors [
7
,
9
,
17
]. The Fc-dependent mechanism of action plays in important role in
amivantamab-vmjw pharmacological activity through antibody-dependent cellular cyto-
toxicity by activation of natural killer cells, macrophages, and monocytes. A subsequent
cytokine and chemokine release is triggered along with downmodulation of EGFR and
MET receptors through trogocytosis with Fc interaction [7].
Amivantamab-vmjw is a bispecific monoclonal antibody produced from two different
bivalent parental antibodies. The EGFR arm is a derivative of zalutumumab, which has a
conformational epitope of EGFR domain III, but is different from epitopes of cetuximab
and panitumumab. The epitope on the MET-binding arm blocks the HGF ligand but is
dissimilar from onartuzumab [
18
]. Amivantamab-vmjw binds to extracellular domains of
EGFR and MET receptors with a dissociation constant of 1.43 and 0.04 nM, respectively [
19
].
3. Pharmacokinetics and Pharmacodynamics
The phase 2 clinical trial recommended dose of amivantamab-vmjw was selected
based on serum EGFR and MET target saturation in addition to the determined preclinical
concentration of 168 ug/mL [
8
]. Saturation of EGFR and MET circulating targets was
achieved with doses above 700 mg. Dosing was determined by weight utilizing population
kinetics to minimize pharmacokinetic variability, leading to recommendations of 1400 mg
with weight
80 kg and 1050 mg for <80 kg. The volume of distribution and clearance
of amivantamab-vmjw increased due to body weight escalation, which is attributed to
the two-tiered weight-based dosing. Exposure was similar when comparing the 1050 mg
dose at <80 kg and 1400 mg for those who weighed
80 kg. Pharmacokinetic results of
amivantamab-vmjw showed no clinically significant differences regarding age sex, race,
Biomedicines 2023,11, 950 3 of 11
creatinine clearance (CrCl)
29 mL/min, or mild hepatic impairment. Amivantamab-
vmjw was not investigated in CrCl < 29 mL/min or in moderate (bilirubin 1.5 to 3 times
upper limit normal (ULN)) or severe (total bilirubin > 3 times ULN) hepatic impairment.
Mean linear clearance was determined to be 0.36 L/d and have a terminal half-life of
11.3 (+/4.53) days [8,15].
4. Clinical Trial Efficacy
The safety and efficacy of amivantamab-vmjw was evaluated in the CHRYSALIS trial,
a phase I, open-label, multicenter, dose escalation and dose expansion study that included
patients with advanced NSCLC with EGFR exon20ins mutation. Patients included were
adults
18 years, had metastatic or unresectable NSCLC, ECOG status of
1, and had
progressed on standard of care therapy. Patients with untreated or active brain metastasis
were not included in the trial. Previously treated and asymptomatic brain metastases
were allowed. Patients in the dose expansion phase had documented EGFR mutations or
MET mutations and quantified disease defined by RECIST 1.1 criteria. Tissue or central
next-generation sequencing (NGS) circulating tumor DNA (ctDNA) testing was utilized to
detect EGFR exon20ins mutations.
The dose escalation phase was a 3 + 3 design that assessed amivantamab-vmjw
dosing weekly for the first 28 days followed by biweekly dosing. Patients were divided
into six different dose cohorts for dose escalation. The analysis presented here includes
patients with NSCLC EGFR exon20ins mutations (cohort D). The main objective of the dose
escalation phase was to determine the maximum tolerated dose for the phase II portion of
the study. For phase II, dose expansion, six cohorts were identified based on EGFR and MET
mutations or amplifications and previous therapy status. The primary objective for the
phase II portion of the study was ORR. Secondary endpoints included DOR; clinical benefit
rate (CBR), defined as PR or CR or stable disease for
11 weeks; PFS; and OS. Treatment
with amivantamab-vmjw extended until disease progression, unacceptable toxicity, or
withdrawal of consent.
From May 2016 to June 2020, 362 patients were enrolled in the study. Patients com-
prised in the safety population included those with EGFR exon20ins mutations that pro-
gressed on platinum-based therapy and were treated at the phase II dose (n = 114).
A total
of 258 patients were given a dosage of 1050 mg for patients weighing < 80 kg and 1400 mg
for patients
80 kg given once weekly for the first four weeks, then biweekly starting at
week five. The efficacy population consisted of the first 81 patients with EGFR exon20ins
mutations that had received previous platinum-based chemotherapy and had
3 disease
assessments at data cutoff. Data were assessed through 8 October 2020 for this popula-
tion. The median age of the efficacy population was 62 years (42–84 years), and 59% were
women, 49% were of Asian descent, 53% were nonsmokers, and 95% had adenocarcinoma.
The median number of previous lines of therapy was two (range 1–7). All patients had
previously received platinum-based chemotherapy, 25% received prior EGFR TKIs, and
46% had received prior immuno-oncology therapies. Approximately 22% of patients had
treated brain lesions at enrollment.
At median follow up of 9.7 months, the ORR was 40% (95% CI, 29–51) with 3 confirmed
CR and 29 PR outcomes, as evaluated by a blinded independent central review. The
investigator-assessed ORR was 36% (95% CI, 25–47). The median DOR was 11.1 months
(95% CI, 6.9-NR). CBR was observed in 74% of patients (95% CI, 63–83). Median PFS was
8.3 months (95% CI, 6.5–10.9). Median OS was 22.8 months (95% CI, 14.6-NR). The overall
survival endpoint is currently immature. Efficacy endpoints are summarized in Table 1[
8
].
Biomedicines 2023,11, 950 4 of 11
Table 1.
Efficacy Outcomes of Amivantamab-vmjw for NSCLC Harboring EGFR Exon20ins
Mutations [8].
Response CR No.
(%)
PR No.
(%)
SD No.
(%)
PD No.
(%)
ORR %
(95% CI)
NE No.
(%)
mPFS
(95% CI)
mOS
(95% CI)
Efficacy
population (n = 81)
3 (4) 29 (36) 39 (48) 8 (10) 40 (29–51) 2 (2) 8.3 mo
(6.5–10.9)
22.8 mo
(14.6-NR)
CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; ORR, overall response
rate; NE, not evaluable; mPFS, median progression-free survival; mo. Months; mOS, median overall survival; NR,
not reached; NSCLC, non-small cell lung cancer.
5. Safety
The National Cancer Institute Common Terminology Criteria for Adverse Events
(CTCAE), version 4.03 was applied for grading AEs. The most frequent AEs observed in the
CHRYSALIS trial were rash (84%), infusion-related reactions (IRR (64%)), paronychia (50%),
musculoskeletal pain (47%), dyspnea (37%), nausea (36%), fatigue (33%), edema (27%),
stomatitis (26%), cough (25%), constipation (23%), and vomiting (22%). Rash including
acneiform dermatitis, paronychia, stomatitis, and diarrhea are associated with on-target
anti-EGFR TKIs activity, while peripheral edema is associated with on-target anti-MET
activity. Grade
3 AEs were reported in 16% of patients including rash (4%), IRRs (3%),
and neutropenia (3%). All grade interstitial lung disease and pneumonitis occurred in 4%
of patients [8,15]. The types and rates of AEs are summarized in Table 2.
Table 2. Reported Adverse Events with Amivantamab-vmjw in CHRYSALIS [8].
Common AE Safety Population (n = 114) No. (%) Patients Treated with the Phase II Dose
(n = 258) No. (%)
Total Grade 1 Grade 2 Grade 3 Total Grade 1 Grade 2 Grade 3
Rash 98 (86) 43 (38) 51 (45) 4 (4) 202 (78) 101 (39) 94 (36) 7 (3)
Infusion-related
reactions 75 (66) 9 (8) 63 (55) 3 (3) 167 (65) 21 (8) 140 (54) 6 (2)
Paronychia 51 (45) 28 (25) 22 (19) 1 (1) 104 (40) 50 (19) 51 (20) 3 (1)
Constipation 27 (24) 18 (16) 9 (8) 0 58 (23) 36 (14) 22 (9) 0
Dyspnea 22 (19) 12 (11) 8 (7) 2 (2) 52 (20) 28 (11) 13 (5) 11 (4)
Nausea 22 (19) 17 (15) 5 (4) 0 (55) 21 40 (16) 14 (5) 1 (0.4)
Vomiting 12 (11) 10 (9) 2 (2) 0 29 (11) 22 (9) 6 (2) 1 (0.4)
Fatigue 21 (18) 15 (13) 4 (4) 2 (2) 47 (18) 29 (11) 16 (6) 2 (1)
Stomatitis 24 (21) 11 (10) 13 (11) 0 50 (19) 33 (13) 17 (7) 0
Cough 16 (14) 11 (10) 5 (4) 0 40 (16) 25 (10) 15 (6) 0
Myalgia 14 (12) 12 (11) 2 (2) 0 28 (11) 23 (9) 5 (2) 0
Dose reduction due to AEs occurred in 13% of patients of which rash was the most
frequent cause (10%). Adverse events causing permanent discontinuation of treatment
occurred in
11% of patients and included pneumonia, IRRs, pneumonitis/interstitial lung
disease, pleural effusion, and rash [8,15].
Overall, 66% of patients treated with amivantamab-vmjw resulted in IRRs, causing in-
fusion interruptions in 59% of patients. IRRs developed almost exclusively on
cycle 1 day 1
(65%) or day 2 (3.4%). IRRs rarely developed during subsequent cycles (1.1%). Of the
reported IRRs, 97% of them were grade 1–2, 2.2% were grade 3, and 0.4% were
grade 4
.
There were no predisposing factors identified. To mitigate the development of IRRs, the
first dose was split over two days (cycle 1 day 1 and 2). Prophylactic premedications were
required (Table 3). The median time for onset was 1 h (range, 0.1 to 18 h) after the start of the
infusion. Dose modifications due to IRRs occurred in 62% of patients, and
1.3% terminated
treatment due to IRRs. Signs and symptoms of IRR are listed in Table 2.
Biomedicines 2023,11, 950 5 of 11
Table 3. Prophylactic Premedications for Infusion-Related Reactions and Antiemetics [8].
Required Premedications
Medication Dose and Route of
Administration
Dosing Window Prior to
Drug Administration Cycle/Day
Glucocorticoid aDexamethasone 10 mg IV or
Methylprednisolone 40 mg IV 45 to 60 min Cycle 1 day 1 and cycle 1
day 2 only
Antihistamine bDiphenhydramine 25–50 mg IV, 15 to 30 min
PO, 30 to 60 min All cycles
Antipyretic bAcetaminophen 650–1000 mg IV, 15 to 30 min
PO, 30 to 60 min All cycles
Optional Premedications
Glucocorticoid aDexamethasone 10 mg or
Methylprednisolone 40 mg
IV, 45 to 60 min
PO, 60 to 90 min Cycle 1 day 8 and beyond
H2 receptor antagonist Famotidine 20 mg IV 15 to 30 min Any cycle
Antiemetics Ondansetron 8 mg PO or 16 mg IV 15 to 30 min Any cycle
PO, oral; IV, intravenous.
a
beginning with cycle 1 day 8, optional pre-dose steroids were administered if clinically
indicated for patients who experienced an infusion-related reaction on cycle 1 day 1 or cycle 1 day 2.
b
Required
at all doses.
If patients experienced infusion reactions during subsequent cycles, infusion was
interrupted until symptom resolution and resumed at 50% of the previous infusion rate [
8
].
Management of IRRs included infusion interruptions, reducing the rate of administration,
and administration of supportive care, including antihistamines, antipyretics, glucocorti-
coids, epinephrine, and meperidine, which is shown in Table 3[8].
EGFR inhibitors have been shown to cause skin-related toxicities [
17
,
20
]. Prevention of
the EGFR inhibitor induced rash-related AEs during the CHRYSALIS trial, which included
using broad-spectrum sunscreen and alcohol-free emollients and limiting exposure to
sunlight [
8
]. Management of EGFR inhibitor-induced rash consisted of antihistamines,
corticosteroids, topical antibiotics (clindamycin), systemic antibiotics (doxycycline 100 mg
BID or minocycline 100 mg BID), antiseptic soaks, and local corticosteroids [8].
6. Dosing and Administration
The suggested dosing for amivantamab-vmjw is based on the patient’s baseline body
weight. Subsequent body weight changes do not require further dose adjustments. For
patients weighing < 80 kg, the recommended dose is 1050 mg (3 vials) [
15
]. For patients
weighing
80 kg, the recommended dose is 1400 mg (4 vials) [
15
]. For the first 4 weeks,
amivantamab-vmjw is administered weekly. Starting at week 5, amivantamab-vmjw is ad-
ministered every 2 weeks (Table 4). Administration of an antihistamine (diphenhydramine
25 to 50 mg) and antipyretic (acetaminophen 650 to 1000 mg) is required prior to all doses
of amivantamab-vmjw. A glucocorticoid (dexamethasone 10 mg or methylprednisolone
40 mg
) administration is mandatory prior to cycle 1, days 1 and 2 of amivantamab-vmjw
infusion (Table 3) [
15
]. Dose modifications for toxicity are outlined in Table 5, and modifica-
tion guidance for adverse reactions is found in Table 6.
Table 4. Amivantamab-vmjw Dose Based on Body Weight and Dosing Schedule [15].
Baseline Body Weight (kg) Dose (mg) Number of 350 mg/7 mL Vials
<80 kg 1050 mg 3
80 kg 1400 mg 4
Dosing Schedule
Cycle 1–4 Week 1 aSplit infusion on C1D1 and C1D2
Weeks 2–4 Infusion on Day 1
Cycles 5+ Every 2 weeks starting at week 5
aC1D1: Cycle 1 Day 1; C1D2: Cycle 1 Day 2.
Biomedicines 2023,11, 950 6 of 11
Table 5. Amivantamab-vmjw Dose Reduction Levels for Adverse Reactions [15].
Body Weight at Baseline Dose Level Dose
Less than 80 kg 0 1050 mg
1 700 mg
2 350 mg
Greater than or equal to 80 kg
3
0
1
2
3
Discontinue
1400 mg
1050 mg
700 mg
Discontinue
Table 6. Amivantamab-vmjw Dose Modifications for Adverse Reactions [15].
Toxicity Severity
(CTCAE Grade *) Dose Modification
Dermatologic
2
Supportive care treatment
Reassess after 2 weeks
Dose reduction if rash does not improve
3
Hold amivantamab
Supportive care management
Resume with dose reduction Grade 2
Discontinue if no improvement within 2 weeks
4Permanently discontinue
Severe blistering, bullous,
exfoliation of skin
(including TENS)
Permanently discontinue
Infusion related
reactions
1 or 2
Stop infusion and monitor for symptom resolution
Supportive care management
Resume at 50% initial infusion rate
Escalate infusion rate after 30 min if no additional symptoms occur
Corticosteroid premedication for subsequent infusions a
3
Stop infusion and monitor for symptom resolution
Supportive care management
Resume at 50% initial infusion rate
Escalate infusion rate after 30 min if no additional symptoms occur
Corticosteroid premedication for subsequent infusions a
Permanently discontinue with recurrent Grade 3
4Permanently discontinue
Interstitial lung dis-
ease/Pneumonitis Any Hold dose if suspected
Permanently discontinue if confirmed
Other 3
Hold dose until recover Grade 1 or baseline
Resume same dose if recovery within 1 week
Resume with dose reduction if recovery within 1–4 weeks
Permanently discontinue if no recovery in 4 weeks
4
Hold dose until recover Grade 1 or baseline
Resume with dose reduction if recovery within 4 weeks
Permanently discontinue if no recovery in 4 weeks
Permanently discontinue with recurrent Grade 4
TENS: toxic epidermal necrolysis. * NCI Common Terminology Criteria for Adverse Events (CTCAE) severity
Grade 1–5 scale.
a
Dexamethasone 10 mg IV or methylprednisolone 40 mg IV 45–60 min prior to amivantamab
administration.
Amivantamb-vmjw is administered as an intravenous infusion using an in-line low
protein-binding polyethersulfone 0.2-micron filter. To minimize infusion reactions, the first
dose is split over day 1 and day 2. Additionally, the administration tubing set is primed
with 15–25 mL of diluent (dextrose or saline) to slow initial exposure to amivantamab-vmjw.
Biomedicines 2023,11, 950 7 of 11
Another strategy to minimize the risk for IRRs is withdrawing and discarding 10 mL of
blood prior to flushing the catheter with dextrose or saline at the end of drug administration
to avoid rapid infusion of residual amivantamab-vmjw. Additionally, do not flush when
infusion must be interrupted for toxicity [21].
While there are no data evaluating amivantamab-vmjw administration vis a peripheral
versus central line, it is suggested to infuse amivantamab-vmjw via a peripheral line in
weeks 1 and 2 when IRRs are most frequent. Administration via a peripheral line allows for
slower infusion, thus reducing the risk for IRRs. A central line can be utilized following the
first two weeks of therapy. It is also recommended to consider dose preparation as close
to administration time as possible to allow for potential extended infusion times if IRRs
occur [
22
]. Recommended administration rates vary based on dose and treatment cycles
and are summarized in Table 7.
Table 7. Infusion Rates of Amivantamab-vmjw Administration [15].
1050 mg Dose (for <80 Kg)
Week Dose (Prepared in 250 mL Bag) Initial Infusion Rate Subsequent Infusion Rate
1 (split dose)
Day 1 350 mg 50 mL/h 75 mL/h
Day 2 700 mg 50 mL/hr 75 mL/h
2 1050 mg 85 mL/h
3+ 1050 mg 125 mL/h
1400 mg Dose (for 80 Kg)
Week Dose (prepared in 250 mL bag) Initial Infusion Rate Subsequent Infusion Rate
1 (split dose)
Day 1 350 mg 50 mL/h 75 mL/h
Day 2 1050 mg 35 mL/h 50 mL/h
2 1400 mg 65 mL/h
3+ 1400 mg 125 mL/h
7. Cost
Amivantamab-vmjw is available as a single-dose 350 mg/7 mL (50 mg/mL) vial. The
average wholesale price for each vial is around USD 3296 [
23
]. For patients weighing less
than 80 kg, the dose is three vials, and the cost is approximately USD 9888. For patients
weighing more than or equal to 80 kg, the dose is four vials, the cost is approximately
USD 13,184
. The CHRYSALIS study patients were treated for the median duration of
5.6 months
, which amounts to approximately USD 138,432 to USD 184,576 for six treatment
cycles. Center for Medicare and Medicaid Services (CMS) has established a new HCPCS
level II code J9061 in 2 mg increments for billing purposes.
8. Application to Clinical Practice
Patients with EGFR exon20ins mutations in NSCLC represent 10–12% of all EGFR
mutations. Prognosis is usually poor, and effective treatment options have been very limited.
Efficacy with EGFR TKIs, such as erlotinib or osimertinib, are limited by low-binding
affinity at the tyrosine kinase domain of Exon 20. Upfront platinum-based chemotherapy
is offset by a median OS of 12.5 months in the relapsed or refractory setting, making
amivantamab-vmjw an attractive choice for second- or third-line therapy [24].
Recently, an oral EGFR exon20ins mutation-directed TKI, mobocertinib, was approved
by the FDA for treatment of advanced NSCLC. The ORR in 114 patients with NSCLC har-
boring EGFR exon20ins mutations who had made progress on platinum-based chemother-
apy was 28% (95% CI, 20–37) [
25
]. Approximately 46% of patients exhibited treatment-
related AEs of grade
3. Diarrhea and rash were the most frequently reported AEs, at
90% (21% grade 3) and 45%, respectively [25].
Amivantamab-vmjw presents a manageable safety and toxicity profile with IRRs being
the most common adverse effect. IRRs can be mitigated with splitting of the dose over
Biomedicines 2023,11, 950 8 of 11
two days
during week 1 and administration of antihistamines, antipyretics, and glucocorti-
coids prior to infusion. IRRs were managed by infusion interruption and reduction of the
infusion rate. The incidence of IRRs decreases dramatically from day 1 to day 2 and in subse-
quent weeks with reported rates of 65% on week 1 day 1, 3.4% on day 2, 0.4% on week 2, and
1.1% for all ensuing infusions. Administration of pre-infusion glucocorticoids is required
on week 1
days 1 and 2
but optional in subsequent cycles. Continuation of pre-infusion
antihistamines and antipyretics is recommended for all cycles. Practical recommenda-
tions include administering amivantamab-vmjw through a peripheral line on
days 1 and 2
,
scheduling infusion appointments early in the morning, preparing amivantamab-vmjw
close to the scheduled administration appointment to allow for extended infusion time
during appropriate working hours of the infusion center, and having cardiopulmonary
resuscitation equipment and medication needed to treat infusion reactions readily available.
Dermatologic reactions were commonly seen in CHRYSALIS trial with 86% of pa-
tients experiencing rash of any grade, including dermatitis acneiform, which is one of
the most common on-target dermatological AEs associated with EGFR inhibitors. The
median time for the onset of rash was 14 days (range, 1 to 276 days). Topical steroids
and/or topical antibiotics and oral antibiotics are typically recommended for EGFR in-
hibitors’ mediated rash. Other interventions include applications of topical emollients
and broad-spectrum UVA/UVB sun lotion during treatment and for 2 months following
discontinuation of treatment.
Amivantamab-vmjw was FDA approved based on early efficacy data that led to a
Breakthrough Therapy designation. This was based on the investigator-assessed ORR
of 41%, median DOR of 7 months, and CBR of 72%. This efficacy analysis is based on a
small sample size of 81 patients [
8
]. Furthermore, patients with active or untreated brain
metastases were excluded, and the benefit of amivantamab-vmjw is unknown in this patient
population, which is a key limitation of this therapy. Efficacy analysis is ongoing for patients
with EGFR exon20ins mutations in other disease states besides NSCLC, in patients who
have not been previously treated with platinum-based chemotherapy, and in combination
with other therapies. The National Comprehensive Cancer Network (NCCN) guidelines
suggest (category 2A) either amivantamab-vmjw or mobocertinib for subsequent therapy
for patients with exon20ins mutations who have progressed during or after initial systemic
treatment including chemotherapy with or without immunotherapy [
26
]. Comparison
of mobocertinib and amivantamab-vmjw is outlined in Table 8. After progression on
amivantamab-vmjw or mobocertinib, the recommendation is to switch to the exon20ins
mutation-targeted treatment that was not previously given. Ongoing clinical trials targeting
exon20ins mutations are included in Table 9.
Table 8.
FDA-Approved Treatments for Locally Advanced or Metastatic NSCLC with EGFR
Exon20ins Mutations in the Recurrent Setting [8].
CHRYSALIS (Amivantamab-Vmjw) (n = 81) Study 101 (Mobocertinib) (n = 114)
ORR 40% (95% CI, 29–51%): 3.7% CR; 36% PR 28% (95% CI, 20–37) all PR
DOR 11.1 mo (95% CI 6.9, NE) 17.5 mo (95% CI, 7.4–20.3; n = 32/114)
DOR 6 mos 63% 59%
mOS 22.8 mo 24 mo
mPFS 8.3 mo 7.3 mo
ORR, overall response rate; CR, complete response; DOR, duration of response; mOS, median overall survival;
mPFS, median progression-free survival; NSCLC, non-small cell lung cancer; PR, partial response.
Biomedicines 2023,11, 950 9 of 11
Table 9. Ongoing Clinical Trials of Exon20ins Mutation-Targeted Therapy in NSCLC [2733].
Trial aInvestigational Intervention(s) Phase Allocation/Design NSCLC Condition
PAPILLON
(NCT04538664)
Amivantamab-vmjw and
carboplatin-pemetrexed compared with
carboplatin-pemetrexed
3Randomized,
open-label EGFR exon20ins mutation
MARIPOSA-2
(NCT04988295)
Amivantamab-vmjw and lazertinib in
combination with platinum-based
chemotherapy compared with
platinum-based chemotherapy alone
3Randomized,
open-label
EGFR-mutated Osimertinib
failure
EXCLAIM
(NCT04129502)
TAK-788 (mobocertinib) compared with
platinum-based chemotherapy 3Randomized,
open-label
EGFR Exon 20 insertion
mutation
CLN-081
(NCT04036682) CLN-081 1/2a N/A, open-label EGFR Exon 20 insertion
mutation
Poziotinib
(NCT03066206) Poziotinib 2 N/A, open-label
Mutant advanced solid tumors
EGFR or HER2
ZENITH20
(NCT03318939) Poziotinib 2 N/A, open-label EGFR or HER2 Exon
20 insertion mutation
FAVOUR
(NCT04858958) Furmonertinib mesilate 1b Randomized,
open-label
EGFR Exon 20 insertion
mutation
aClinicaltrials.gov identifier.
9. Conclusions
The FDA approval of amivantamab-vmjw, the first bispecific antibody to target EGRF
exon20ins, represents an important development in the treatment of patients with advanced
or metastatic NSCLC who have previously had limited effective treatment options. The
early findings of the CHRYSALIS trial not only show a benefit in overall response rate, but
also a relatively tolerable safety and toxicity profile. In addition, the efficacy results show
durable responses in patients who do benefit from this therapy. CHRYSALIS is an ongoing
study that is evaluating the effectiveness and safety of amivantamab-vmjw as monotherapy
and in combination with other therapies in NSCLC and other solid malignancies harboring
EGFR exon20ins mutations. Long-term follow up is necessary to confirm efficacy and safety
in the study population and in real-world patients.
Author Contributions:
Conceptualization and methodology, V.S., A.M. and F.R.; writing-original
draft preparation, all authors; writing-review and editing, all authors. All authors have read and
agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
König, D.; Savic Prince, S.; Rothschild, S.I. Targeted therapy in advanced and metastatic non-small cell lung cancer. An update on
treatment of the most important actionable oncogenic driver alterations. Cancers 2021,13, 804. [CrossRef] [PubMed]
2.
Centers for Disease Control and Prevention. An Update on Cancer Deaths in the United States. Available online: https://www.cdc.
gov/cancer/dcpc/research/update-on-cancer-deaths/ (accessed on 8 February 2022).
3.
American Lung Association. Lung Cancer Fact Sheet. Available online: https://www.lung.org/lung-health-diseases/lung-
disease-lookup/lung-cancer/resource-library/lung-cancer-fact-sheet (accessed on 8 February 2022).
4.
Melosky, B.; Wheatley-Price, P.; Juergens, R.A.; Sacher, A.; Leighl, N.B.; Tsao, M.-S.; Cheema, P.; Snow, S.; Liu, G.; Card, P.B.;
et al. The rapidly evolving landscape of novel targeted therapies in advanced non-small cell lung cancer. Lung Cancer
2021
,160,
136–151. [CrossRef] [PubMed]
5.
Meador, C.B.; Sequist, L.V.; Piotrowska, Z. Targeting EGFR Exon 20 insertions in non-small cell lung cancer: Recent advances and
clinical updates. Cancer Discov. 2021,11, 2145–2157. [CrossRef] [PubMed]
Biomedicines 2023,11, 950 10 of 11
6.
Passaro, A.; Mok, T.; Peters, S.; Popat, S.; Ahn, M.J.; de Marinis, F. Recent advances on the role of EGFR tyrosine kinase inhibitors
in the management of NSCLC with uncommon, non exon 20 insertions, EGFR Mutations. J. Thorac. Oncol.
2021
,16, 764–773.
[CrossRef] [PubMed]
7.
Vijayaraghavan, S.; Lipfert, L.; Chevalier, K.; Bushey, B.S.; Henley, B.; Lenhart, R.; Sendecki, J.; Beqiri, M.; Millar, H.J.;
Packman, K.; et al.
Amivantamab-vmjw (JNJ-61186372), an Fc enhanced EGFR/cMet bispecific antibody, induces receptor down-
modulation and antitumor activity by monocyte/macrophage trogocytosis. Mol. Cancer Ther.
2020
,19, 2044–2056. [CrossRef]
[PubMed]
8.
Park, K.; Haura, E.B.; Leighl, N.B.; Mitchell, P.; Shu, C.A.; Girard, N.; Viteri, S.; Han, J.-Y.; Kim, S.-W.; Lee, C.K.; et al. Amivantamab-
vmjw in EGFR exon 20 insertion-mutated non-small-cell lung cancer progressing on platinum chemotherapy: Initial results from
the CHRYSALIS phase I study. J Clin. Oncol. 2021,39, 3391–3402. [CrossRef] [PubMed]
9.
Yun, J.; Lee, S.H.; Kim, S.Y.; Jeong, S.Y.; Kim, J.H.; Pyo, K.H.; Park, C.W.; Heo, S.G.; Yun, M.R.; Lim, S.; et al. Antitumor activity
of amivantamab-vmjw (JNJ-61186372), an EGFR-MET bispecific antibody, in diverse models of EGFR exon 20 insertion-driven
NSCLC. Cancer Discov. 2020,10, 1194–1209. [CrossRef] [PubMed]
10.
Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.;
Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med.
2020,382, 41–50. [CrossRef]
11.
Beau-Faller, M.; Prim, N.; Ruppert, A.M.; Nanni-Metéllus, I.; Lacave, R.; Lacroix, L.; Escande, F.; Lizard, S.; Pretet, J.-L.;
Rouquette, I.; et al.
Rare EGFR exon 18 and exon 20 mutations in non-small-cell lung cancer on 10 117 patients: A multicentre
observational study by the French ERMETIC-IFCT network. Ann. Oncol. 2014,25, 126–131. [CrossRef] [PubMed]
12.
Naidoo, J.; Sima, C.S.; Rodriguez, K.; Busby, N.; Nafa, K.; Ladanyi, M.; Riely, G.J.; Kris, M.G.; Arcila, M.E.; Yu, H.A. Epidermal
growth factor receptor exon 20 insertions in advanced lung adenocarcinomas: Clinical outcomes and response to erlotinib. Cancer
2015,121, 3212–3220. [CrossRef] [PubMed]
13.
Yasuda, H.; Park, E.; Yun, C.H.; Sng, N.J.; Lucena-Araujo, A.R.; Yeo, W.-L.; Huberman, M.S.; Cohen, D.W.; Nakayama,
S.;
Ishioka, K.; 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, Erratum in Sci. Transl. Med.
2014
,6, 225er1. [CrossRef]
[PubMed]
14.
Lee, C.K.; Davies, L.; Wu, Y.L.; Mitsudomi, T.; Inoue, A.; Rosell, R.; Zhou, C.; Nakagawa, K.; Thongprasert, S.; Fukuoka, M.; et al.
Gefitinib or Erlotinib vs Chemotherapy for EGFR Mutation-Positive Lung Cancer: Individual Patient Data Meta-Analysis of
Overall Survival. J. Natl. Cancer Inst. 2017,109, djw279. [CrossRef] [PubMed]
15. Horsham, P.A. Amivantamab-Vmjw (Rybrevant) [Package Insert]; Janssen Biotech: Horsham Township, PA, USA, 2021.
16.
Zhang, Z.; Yang, S.; Wang, Q. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-
mutant lung cancer. Biomark. Res. 2019,7, 27. [CrossRef] [PubMed]
17. Kozuki, T. Skin problems and EGFR-tyrosine kinase inhibitor. Jpn. J. Clin. Oncol. 2016,46, 291–298. [CrossRef] [PubMed]
18.
Neijssen, J.; Cardoso, R.M.F.; Chevalier, K.M.; Wiegman, L.; Valerius, T.; Anderson, G.M.; Moores, S.L.; Schuurman, J.;
Parren, P.W.
;
Strohl, W.R.; et al. Discovery of amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR and MET. J. Biol. Chem.
2021
,
296, 100641. [CrossRef] [PubMed]
19.
Moores, S.L.; Chiu, M.L.; Bushey, B.S.; Chevalier, K.; Luistro, L.; Dorn, K.; Brezski, R.J.; Haytko, P.; Kelly, T.; Wu, S.-J.; et al.
A Novel
Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor-Resistant Lung Tumors. Cancer Res.
2016,76, 3942–3953. [CrossRef] [PubMed]
20.
Giovannini, M.; Gregorc, V.; Belli, C.; Roca, E.; Lazzari, C.; Viganò, M.G.; Serafico, A.; Villa, E. Clinical Significance of Skin Toxicity
due to EGFR-Targeted Therapies. J. Oncol. 2009,2009, 849051. [CrossRef] [PubMed]
21.
Data on File. Investigational Product Preparation Instruction for Intravenous Administration of JNJ-61186372 (Amivantamab) for Global
Sites; TV-TEC-168067; Janssen Research & Development, LLC.: Raritan, NJ, USA, 2021.
22.
Park, K.; Sabari, J.; Haura, E.B.; Shu, A.; Spira, R.; Salgia, K.L.; Reckamp, R.E.; Sanborn, R.; Govindan, J.M.; Bauml, J.C.; et al.
Management of Infusion-Related Reactions (IRRs) in Patients Receiving Amivantamab. In Proceedings of the European Society
for Medical Oncology (ESMO) Virtual Congress 2021, Webcast Online, 16–21 September 2021.
23.
Drugs.com. Rybrevant Prices, Coupons and Patient Assistance Programs. Available online: https://www.drugs.com/price-
guide/rybrevant (accessed on 29 January 2022).
24.
Dersarkissian, M.; Bhak, R.; Lin, H.; Li, S.; Cheng, M.; Lax, A.; Huang, H.; Duh, M.; Ou, S. P2.01-103 Real-world treatment
patterns and survival in non-small cell lung cancer patients with EGFR exon 20 insertion mutations. J. Thorac. Oncol.
2019
,
14, S681. [CrossRef]
25.
Zhou, C.; Ramalingam, S.S.; Kim, T.M.; Kim, S.W.; Yang, J.C.-H.; Riely, G.J.; Mekhail, T.; Nguyen, D.; Campelo, M.R.G.;
Felip, E.; et al.
Treatment outcomes and safety of mobocertinib in platinum-pretreated patients with EGFR exon 20 insertion-
positive metastatic non-small cell lung cancer: A phase 1/2 open-label nonrandomized clinical trial. JAMA Oncol.
2021
,7, e214761.
[CrossRef] [PubMed]
26.
National Comprehensive Cancer Network. Non-Small Cell Lung Cancer (Version 3.2022). Available online: https://www.nccn.
org/professionals/physician_gls/pdf/nscl.pdf (accessed on 11 July 2022).
Biomedicines 2023,11, 950 11 of 11
27.
Janssen Research & Development. A Study of Combination Amivantamab and Carboplatin-Pemetrexed Therapy, Compared
with Carboplatin-Pemetrexed, in Participants with Advanced or Metastatic Non-Small Cell Lung Cancer Characterized by
Epidermal Growth Factor Receptor (EGFR) Exon 20 Insertions (PAPILLON). ClinicalTrials.gov Identifier: NCT04538664. Updated
21 June 2022. Available online: https://www.clinicaltrials.gov/ct2/show/record/NCT04538664 (accessed on 29 June 2021).
28.
Janssen Research & Development, LLC. A Study of Amivantamab and Lazertinib in Combination with Platinum-Based
Chemotherapy Compared with Platinum-Based Chemotherapy in Patients with Epidermal Growth Factor Receptor (EGFR)-
Mutated Locally Advanced or Metastatic Non- Small Cell Lung Cancer after Osimertinib Failure (MARIPOSA-2). ClinicalTri-
als.gov Identifier: NCT04988295. Updated 21 June 2022. Available online: https://www.clinicaltrials.gov/ct2/show/record/
NCT04988295 (accessed on 29 June 2021).
29.
Takeda. TAK-788 as First-Line Treatment Versus Platinum-Based Chemotherapy for Non-Small Cell Lung Cancer (NSCLC)
with EGFR Exon 20 Insertion Mutations. ClinicalTrials.gov Identifier: NCT04129502. Updated 21 June 2022. Available online:
https://www.clinicaltrials.gov/ct2/show/record/NCT04129502 (accessed on 29 June 2021).
30.
Cullinan Pearl. A Phase 1/2a, Open-Label, Multi-Center Trial to Assess Safety, Tolerability, Pharmacokinetics, Pharmacody-
namics, and Efficacy of CLN-081 in Patients with Non-Small Cell Lung Cancer Harboring EGFR Exon 20 Insertion Mutations.
ClinicalTrials.gov Identifier: NCT04036682. Updated 21 June 2022. Available online: https://www.clinicaltrials.gov/ct2/show/
record/NCT04036682 (accessed on 29 June 2021).
31.
M.D. Anderson Cancer Center. Poziotinib in EGFR Exon 20 Mutant Advanced NSCLC. ClinicalTrials.gov Identifier:
NCT03066206. Updated 21 June 2022. Available online: https://www.clinicaltrials.gov/ct2/show/record/NCT03066206
(accessed on 29 June 2021).
32.
Spectrum Pharmaceuticals, Inc. Phase 2 Study of Poziotinib in Patients With NSCLC Having EGFR or HER2 Exon 20 Insertion
Mutation. ClinicalTrials.gov Identifier: NCT03318939. Updated 21 June 2022. Available online: https://www.clinicaltrials.gov/
ct2/show/record/NCT03318939 (accessed on 29 June 2021).
33.
Allist Pharmaceuticals, Inc. Study of FURMONERTINIB in Patients with NSCLC Having Exon 20 Insertion Mutation (FAVOUR).
ClinicalTrials.gov Identifier: NCT04858958. Updated 21 June 2022. Available online: https://www.clinicaltrials.gov/ct2/show/
record/NCT04858958 (accessed on 29 June 2021).
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Amivantamab (JNJ-61186372) (7,15,(25)(26)(27) is a bispecific antibody targeting EGFR and MET, exhibiting immune celldirected activity. Due to its promising efficacy and safety profile, on May 21, 2021, the United States Food and Drug Administration (FDA) granted approval for its use in the treatment of EGFR-exon20ins in platinum-based chemotherapy-experienced or treatment-naïve patients with metastatic NSCLC (28,29). ...
Article
Full-text available
Epidermal growth factor receptor (EGFR) exon 20 insertion (ex20ins) mutations are the third most prevalent mutation in non-small cell lung cancer (NSCLC), following the 19del and L858R mutations. The unique nature of the EGFR ex20ins mutation poses challenges for the effectiveness of first- and second-generation EGFR tyrosine kinase inhibitors (TKIs). As a result, chemotherapy remains the primary and more effective treatment approach. However, with advancements in time and technology, numerous experimental studies have revealed the potential of novel drugs and therapies to have stronger inhibitory effects on EGFR ex20ins mutations. In this comprehensive review, we provide an overview of the current treatment landscape, recent advancements, and the prospects for patients with advanced NSCLC characterized by EGFR ex20ins mutations.
... and overall survival (OS) was 22.8 months (95% CI, 14.6-NR). 2 The approval of mobocertinib (Exkivity) was based on data from Study AP32788-15-101 (NCT02716116). The overall response rate (ORR) in 114 patients whose disease had progressed on or after platinum-based chemotherapy was 28% (95% CI: 20%, 37%) with a median duration of response of 17.5 months (95% CI: 7.4, 20.3). ...
Article
Full-text available
Lung cancer is the leading cause of cancer-related deaths worldwide in both men and women. The epidermal growth factor receptor gene exon 20 insertion (EGFRex20ins) mutations are an uncommon and heterogeneous group, resistant to conventional EGFR tyrosine kinase inhibitors (TKIs) and have been considered as an “undruggable target” for a long time. The aim of this scientific paper is to present the data from clinical trials, available in 2021 and confirmed the accelerated approvals for mobocertinib and amivantamab, including some pharmacokinetic and pharmacodynamic characteristics of both medications. We also present the data from PAPILLON study (NCT04538664) that met its primary endpoint with a statistically significant and clinically improvement in progression free survival (PFS) (as measured by BICR) in patients receiving amivantamab (Rybrevant). In 2021, two medications amivantamab and mobocertinib received accelerated approval for treatment of advanced NSCLC with EGFRex20ins mutations. Both medications had shown promising results and similar efficacy in the early phase clinical trials. However, in 2023, after collection additional data for efficacy, the FDA and Takeda have decided to voluntarily withdraw Exkivity from the global market. Janssen Pharmaceutical submitted a supplemental Biologics License Application (sBLA) to the FDA seeking the expanded approval of Rybrevant in combination with chemotherapy. Keywords: Non-small cell lung cancer (NSCLC), epidermal growth factor receptor gene exon 20 insertion mutations (EGFRex20ins) mutation, Amivantamab, Mobocertinib
... 9 Amongst those, early monovalent and bivalent mAb approaches relied on antagonism and antibodydependent cellular cytotoxicity (ADCC) as the main modes of action and failed in clinical development. [10][11][12] Nextgeneration biologics were designed as bispecific antibodies, such as amivantamab 13 or MM-131, 14 or focused on target degradation. [15][16][17] One molecular mechanism for effective target degradation is crosslinking of more than two target molecules via dual binding of suitable combinations of paratopes and their optimal orientation in a biparatopic antibody to favor inter-over intra-molecular binding ( Figure 1a). ...
Article
Full-text available
Optimal combinations of paratopes assembled into a biparatopic antibody have the capacity to mediate high-grade target cross-linking on cell membranes, leading to degradation of the target, as well as antibody and payload delivery in the case of an antibody-drug conjugate (ADC). In the work presented here, molecular docking suggested a suitable paratope combination targeting c-MET, but hydrophobic patches in essential binding regions of one moiety necessitated engineering. In addition to rational design of HCDR2 and HCDR3 mutations, site-specific spiking libraries were generated and screened in yeast and mammalian surface display approaches. Comparative analyses revealed similar positions amendable for hydrophobicity reduction, with a broad combinatorial diversity obtained from library outputs. Optimized variants showed high stability, strongly reduced hydrophobicity, retained affinities supporting the desired functionality and enhanced producibility. The resulting biparatopic anti-c-MET ADCs were comparably active on c-MET expressing tumor cell lines as REGN5093 exatecan DAR6 ADC. Structural molecular modeling of paratope combinations for preferential inter-target binding combined with protein engineering for manufacturability yielded deep insights into the capabilities of rational and library approaches. The methodologies of in silico hydrophobicity identification and sequence optimization could serve as a blueprint for rapid development of optimal biparatopic ADCs targeting further tumor-associated antigens in the future.
Article
Full-text available
The complex nature of cancer cells poses a challenge to the scientific community [...]
Article
Full-text available
The aim of this study is to assess the conservative treatment of acute solitary caecal diverticulitis. The antimicrobial therapy of caecal diverticulitis is reported as successful. The inflammatory process of the cecal diverticulum has been reduced and reverse regression of inflammatory signs was observed.
Article
Full-text available
Background Epidermal growth factor receptor (EGFR) exon 20 insertion (ex20ins) mutation is the third most common EGFR-mutant form, accounting for 10–12% of all EGFR mutations in non-small cell lung cancer (NSCLC). Chemotherapy was the first-line treatment for patients with EGFR ex20ins mutation in the era when EGFR ex20ins tyrosine kinase inhibitors (EGFR ex20ins-TKIs) were inaccessible. Although EGFR ex20ins-TKIs have since then demonstrated certain efficacy, the population benefit rate is not high due to the high cost of the drug and limited benefit to the population. Therefore, the choice of treatment modality when a patient does not have access to EGFR ex20ins-TKIs or are resistant to them remains an avenue worth exploring. Case Description In this report, we present two cases of patients with lung adenocarcinoma and EGFR ex20ins mutation. The two patients were middle-aged Asian women with no smoking history, and both had one or more metastatic lesions. Both achieved long-term clinical benefit (progression-free survival ≥12 months) after receiving combined treatment, suggesting that this is a promising treatment modality. Conclusions To the best of our knowledge, this is the first report supporting the combination of stereotactic body radiotherapy and apatinib and camrelizumab as an effective treatment strategy in patients with advanced EGFR ex20ins-positive NSCLC who have been previously treated with chemotherapy. The therapy described in this report might serve as a potential alternative approach for clinical oncologists.
Article
Full-text available
Importance: Metastatic non-small cell lung cancer (mNSCLC) with EGFR exon 20 insertion (EGFRex20ins) mutations is associated with a poor prognosis. Mobocertinib is an oral tyrosine kinase inhibitor designed to selectively target EGFRex20ins mutations. Objective: To evaluate treatment outcomes and safety of mobocertinib in patients with previously treated EGFRex20ins-positive mNSCLC. Design, setting, and participants: This 3-part, open-label, phase 1/2 nonrandomized clinical trial with dose-escalation/dose-expansion cohorts (28 sites in the US) and a single-arm extension cohort (EXCLAIM; 40 sites in Asia, Europe, and North America) was conducted between June 2016 and November 2020 (data cutoff date). The primary analysis populations were the platinum-pretreated patients (PPP) cohort and the EXCLAIM cohort. The PPP cohort included 114 patients with platinum-pretreated EGFRex20ins-positive mNSCLC who received mobocertinib 160 mg once daily from the dose-escalation (n = 6), dose-expansion (n = 22), and EXCLAIM (n = 86) cohorts. The EXCLAIM cohort included 96 patients with previously treated EGFRex20ins-positive mNSCLC (10 were not platinum pretreated and thus were excluded from the PPP cohort). Interventions: Mobocertinib 160 mg once daily. Main outcomes and measures: The primary end point of the PPP and EXCLAIM cohorts was confirmed objective response rate (ORR) assessed by independent review committee (IRC). Secondary end points included confirmed ORR by investigator, duration of response, progression-free survival, overall survival, and safety. Results: Among the PPP (n = 114) and EXCLAIM (n = 96) cohorts, the median (range) age was 60 (27-84) and 59 (27-80) years, respectively; most patients were women (75 [66%] and 62 [65%], respectively) and of Asian race (68 [60%] and 66 [69%], respectively). At data cutoff, median follow-up was 14.2 months in the PPP cohort (median 2 prior anticancer regimens; 40 [35%] had baseline brain metastases), with confirmed ORR of 28% (95% CI, 20%-37%) by IRC assessment and 35% (95% CI, 26%-45%) by investigator assessment; median duration of response by IRC assessment was 17.5 months (95% CI, 7.4-20.3). Median progression-free survival by IRC assessment was 7.3 months (95% CI, 5.5-9.2). Median overall survival was 24.0 months (95% CI, 14.6-28.8). In the EXCLAIM cohort, median follow-up was 13.0 months, with confirmed ORR by IRC assessment of 25% (95% CI, 17%-35%) and by investigator assessment of 32% (95% CI, 23%-43%). The most common treatment-related adverse events were diarrhea and rash. Conclusions and relevance: In this open-label, phase 1/2 nonrandomized clinical trial, mobocertinib was associated with clinically meaningful benefit in patients with previously treated EGFRex20ins-positive mNSCLC, with a manageable safety profile. Trial registration: ClinicalTrials.gov Identifier: NCT02716116.
Article
Full-text available
Purpose: Non-small-cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) exon 20 insertion (Exon20ins) mutations exhibits inherent resistance to approved tyrosine kinase inhibitors. Amivantamab, an EGFR-MET bispecific antibody with immune cell-directing activity, binds to each receptor's extracellular domain, bypassing resistance at the tyrosine kinase inhibitor binding site. Methods: CHRYSALIS is a phase I, open-label, dose-escalation, and dose-expansion study, which included a population with EGFR Exon20ins NSCLC. The primary end points were dose-limiting toxicity and overall response rate. We report findings from the postplatinum EGFR Exon20ins NSCLC population treated at the recommended phase II dose of 1,050 mg amivantamab (1,400 mg, ≥ 80 kg) given once weekly for the first 4 weeks and then once every 2 weeks starting at week 5. Results: In the efficacy population (n = 81), the median age was 62 years (range, 42-84 years); 40 patients (49%) were Asian, and the median number of previous lines of therapy was two (range, 1-7). The overall response rate was 40% (95% CI, 29 to 51), including three complete responses, with a median duration of response of 11.1 months (95% CI, 6.9 to not reached). The median progression-free survival was 8.3 months (95% CI, 6.5 to 10.9). In the safety population (n = 114), the most common adverse events were rash in 98 patients (86%), infusion-related reactions in 75 (66%), and paronychia in 51 (45%). The most common grade 3-4 adverse events were hypokalemia in six patients (5%) and rash, pulmonary embolism, diarrhea, and neutropenia in four (4%) each. Treatment-related dose reductions and discontinuations were reported in 13% and 4% of patients, respectively. Conclusion: Amivantamab, via its novel mechanism of action, yielded robust and durable responses with tolerable safety in patients with EGFR Exon20ins mutations after progression on platinum-based chemotherapy.
Article
Full-text available
Lung cancer is a highly heterogeneous disease often driven by well-characterized driver mutations. Although the best studied are common alterations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) oncogenes, rapid advances in molecular characterization has led to the development of novel therapeutics that inhibit additional oncogenic alterations in advanced NSCLC. The literature search identified 62 eligible phase I/II clinical trials or integrated analyses of assessing novel targeted agents against the following molecular alterations: ROS1-rearranged, BRAF V600E-mutant, NTRK-rearranged, MET-altered, uncommon EGFR-mutant, RET-rearranged, HER2-positive, KRAS G12C-mutant and NRG1-rearranged. This rapidly evolving field has produced many new targeted treatment options and promising outcomes have led to the FDA approval of seven novel agents for use in ROS1-rearranged, BRAF V600E-mutant, NTRK-rearranged, MET exon 14 skipping-mutant or RET-rearranged advanced NSCLC. Research continues at a rapid pace, with a number of phase III trials underway to fully evaluate new promising agents under development for improving outcomes in patients with NSCLC harboring distinct molecular subtypes. This review will provide a comprehensive summary of existing data as well as a user-friendly guide on the current status of novel targeted therapy in oncogene-driven advanced NSCLC.
Article
Full-text available
A bispecific antibody (BsAb) targeting the epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) pathways represents a novel approach to overcome resistance to targeted therapies in patients with non-small cell lung cancer. In this study, we sequentially screened a panel of BsAbs in a combinatorial approach to select the optimal bispecific molecule. The BsAbs were derived from different EGFR and MET parental monoclonal antibodies (mAbs). Initially, molecules were screened for EGFR and MET binding on tumor cell lines and lack of agonistic activity towards MET. Hits were identified and further screened based on their potential to induce untoward cell proliferation and cross-phosphorylation of EGFR by MET via receptor colocalization in the absence of ligand. After the final step, we selected the EGFR and MET arms for the lead BsAb and added low fucose Fc engineering to generate amivantamab (JNJ-61186372). The crystal structure of the anti-MET Fab of amivantamab bound to MET was solved and elucidated the interaction between the two molecules in atomic details. Amivantamab antagonized the hepatocyte growth factor (HGF)-induced signaling by binding to MET Sema domain and thereby blocking HGF β-chain - Sema engagement. The amivantamab EGFR epitope was mapped to EGFR domain III and residues K443, K465, I467, and S468. Furthermore, amivantamab showed superior antitumor activity over small molecule EGFR and MET inhibitors in the HCC827-HGF in vivo model. Based on its unique mode of action, amivantamab may provide benefit to patients with malignancies associated with aberrant EGFR and MET signaling.
Article
Full-text available
Simple Summary The treatment of advanced and metastatic non-small cell lung cancer (NSCLC) has changed dramatically in recent years due to advanced molecular diagnostics and the recognition of targetable oncogenic driver alterations. This has led to the development of very effective new targeted agents, and thus to a relevant progress in the treatment of oncogene-addicted NSCLC. While the treatment of EGFR-mutated and ALK-rearranged NSCLC is well-established, new targeted therapy options have emerged for other oncogenic alterations. In this comprehensive review article, we discuss the major molecular alterations in NSCLC and the corresponding therapeutic options. Abstract Due to groundbreaking developments and continuous progress, the treatment of advanced and metastatic non-small cell lung cancer (NSCLC) has become an exciting, but increasingly challenging task. This applies, in particular, to the subgroup of NSCLC with oncogenic driver alterations. While the treatment of epidermal growth factor receptor (EGFR)-mutated and anaplastic lymphoma kinase (ALK)-rearranged NSCLC with various tyrosine kinase inhibitors (TKIs) is well-established, new targets have been identified in the last few years and new TKIs introduced in clinical practice. Even for KRAS mutations, considered for a long time as an “un-targetable” alteration, promising new drugs are emerging. The detection and in-depth molecular analysis of resistance mechanisms has further fueled the development of new therapeutic strategies. The objective of this review is to give a comprehensive overview on the current landscape of targetable oncogenic alterations in NSCLC.
Article
Full-text available
The first-line treatment of choice for patients with epidermal growth factor receptor (EGFR) mutation-positive non-small cell lung cancer (NSCLC) is an EGFR tyrosine kinase inhibitor, of which five are predominantly available in practice: gefitinib, erlotinib, afatinib, dacomitinib and osimertinib. The vast majority of prospective clinical trial data with these agents is limited to patients with the common activating and sensitizing EGFR mutations: exon 19 deletions and exon 21 L858R point mutations. However, 10%‒20% of NSCLC patients harbor uncommon EGFR mutations which have variable sensitivity to different EGFR TKIs. Owing to their molecular structures, afatinib, dacomitinib and osimertinib have broader inhibitory profiles than the first-generation agents, gefitinib and erlotinib. Nevertheless, the paucity of prospective clinical data, the wide heterogeneity of uncommon mutations and the existence of compound mutations in up to 25% of cases complicate treatment decisions in this patient subgroup. Here, we collate the latest preclinical and clinical data regarding the activity of different TKIs against major uncommon EGFR mutations including compound mutations, but excluding exon 20 insertions which are generally insensitive to TKIs. Based on these data, we offer suggestions regarding treatment strategies for uncommon EGFR mutations. Moving forward, it will be important to include uncommon EGFR mutations in the first-line molecular analysis of all patients with adenocarcinoma of the lung, as this will help optimize patient outcomes according to their precise genotype.
Article
Full-text available
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 bispecific antibody targeting EGFR–MET, has shown preclinical activity in TKI-sensitive EGFR-mutated NSCLC models and in an ongoing first-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 IFNγ secretion in various models. Importantly, in vivo efficacy 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 preclinical work. These findings 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. This article is highlighted in the In This Issue feature, p. 1079
Article
Approximately 10% of EGFR-activating mutations occur as in-frame insertion mutations in exon 20 of the EGFR kinase domain (EGFR ins20). EGFR ins20 mutations have not demonstrated the same sensitivity to early generations of EGFR tyrosine kinase inhibitors (TKI) as canonical activating EGFR mutations such as del19 and L858R. Development of effective therapies for this subset of patients has been challenging, but recent years have seen more rapid progress in these efforts. In this review, we describe the molecular and clinicopathologic features of EGFR ins20 mutations and summarize recent data on emerging therapies for patients with this subtype of EGFR-mutant non–small cell lung cancer (NSCLC). Significance When activating mutations in EGFR were first discovered in lung cancer, the lack of sensitivity of tumors harboring EGFR ins20 mutations to early-generation EGFR TKIs resulted in this subset of EGFR-mutant tumors being initially classified as an untargetable or intrinsically resistant subpopulation. In addition, the diversity of mutations within EGFR exon 20 and resultant challenges identifying them on routine clinical genotyping tests led to underestimation of their frequency. However, recent scientific progress in targeting EGFR ins20 mutations as well as more effective identification of this clinical cohort has enhanced our ability to develop effective therapies for patients with this subtype of EGFR-mutant NSCLC.
Article
Small molecule inhibitors targeting mutant epidermal growth factor receptor (EGFR) are standard of care in non-small cell lung cancer (NSCLC), but acquired resistance invariably develops through mutations in EGFR or through activation of compensatory pathways such as cMet. Amivantamab (JNJ-61186372) is an anti-EGFR and anti-cMet bispecific low fucose antibody with enhanced Fc function designed to treat tumors driven by activated EGFR and/or cMet signaling. Potent in vivo anti-tumor efficacy is observed upon amivantamab treatment of human tumor xenograft models driven by mutant activated EGFR and this activity is associated with receptor downregulation. Despite these robust anti-tumor responses in vivo, limited anti-proliferative effects and EGFR/cMet receptor downregulation by amivantamab were observed in vitro. Interestingly, in vitro addition of isolated human immune cells notably enhanced amivantamab-mediated EGFR and cMet downregulation leading to antibody dose-dependent cancer cell killing. Through a comprehensive assessment of the Fc-mediated effector functions, we demonstrate that monocytes and/or macrophages, through trogocytosis, are necessary and sufficient for Fc interaction-mediated EGFR/cMet downmodulation and are required for in vivo anti-tumor efficacy. Collectively, our findings represent a novel Fc-dependent macrophage-mediated anti-tumor mechanism of amivantamab and highlight trogocytosis as an important mechanism of action to exploit in designing new antibody-based cancer therapies.