ArticlePDF AvailableLiterature Review

When the MET receptor kicks in to resist targeted therapies

Authors:
  • Institut OncoLille

Abstract and Figures

Although targeted therapies have increased the life expectancy of patients with druggable molecular alterations directly involved in tumor development, the efficacy of these therapies is limited by acquired resistances leading to treatment failure. Most targeted therapies, including ones exploiting therapeutic antibodies and kinase inhibitors, are directed against receptor tyrosine kinases (RTKs) or major signaling hubs. Resistances to these therapies arise when inhibition of these targets is bypassed through activation of alternative signaling pathways. In recent years, activation of the receptor tyrosine kinase MET has been shown to promote resistance to various targeted therapies. This casts MET as important actor in resistance. In this review, we describe how the MET receptor triggers resistance to targeted therapies against RTKs such as EGFR, VEGFR, and HER2 and against signaling hubs such as BRAF. We also describe how MET can be its own resistance factor, as illustrated by on-target resistance of lung tumors harboring activating mutations causing MET exon 14 skipping. Interestingly, investigation of all these situations reveals functional physiological relationships between MET and the target of the therapy to which the cancer becomes resistant, suggesting that resistance stems from preexisting mechanisms. Identification of MET as a resistance factor opens the way to co-treatment strategies that are being tested in current clinical trials.
Content may be subject to copyright.
Oncogene (2021) 40:40614078
https://doi.org/10.1038/s41388-021-01835-0
REVIEW ARTICLE
When the MET receptor kicks in to resist targeted therapies
Marie Fernandes1Philippe Jamme1Alexis B. Cortot1,2 Zoulika Kherrouche1David Tulasne 1
Received: 15 February 2021 / Revised: 26 April 2021 / Accepted: 7 May 2021 / Published online: 24 May 2021
© The Author(s), under exclusive licence to Springer Nature Limited 2021
Abstract
Although targeted therapies have increased the life expectancy of patients with druggable molecular alterations directly
involved in tumor development, the efcacy of these therapies is limited by acquired resistances leading to treatment failure.
Most targeted therapies, including ones exploiting therapeutic antibodies and kinase inhibitors, are directed against receptor
tyrosine kinases (RTKs) or major signaling hubs. Resistances to these therapies arise when inhibition of these targets is
bypassed through activation of alternative signaling pathways. In recent years, activation of the receptor tyrosine kinase
MET has been shown to promote resistance to various targeted therapies. This casts MET as important actor in resistance. In
this review, we describe how the MET receptor triggers resistance to targeted therapies against RTKs such as EGFR,
VEGFR, and HER2 and against signaling hubs such as BRAF. We also describe how MET can be its own resistance factor,
as illustrated by on-target resistance of lung tumors harboring activating mutations causing MET exon 14 skipping.
Interestingly, investigation of all these situations reveals functional physiological relationships between MET and the target
of the therapy to which the cancer becomes resistant, suggesting that resistance stems from preexisting mechanisms.
Identication of MET as a resistance factor opens the way to co-treatment strategies that are being tested in current clinical
trials.
Targeted therapies and the inevitable
associated resistances
Detection of genetic alterations in cancers, supported
mainly by improved genome sequencing, have led to the
discovery of activated oncogenes associated with tumor-
igenesis. These notably include genes encoding actors of
signaling pathways, such as receptor tyrosine kinases
(RTKs), and signaling hubs such as KRAS, BRAF, and
PI3K. Importantly, some tumors depend on a single
deregulated oncogene for their growth and/or survival. This
dependence, called oncogene addiction, explains the ef-
cacy of targeted therapies. The rst example of this was
activation of the BCRABL gene in chronic myelogenous
leukemia, and therapies targeting the ABL kinase have led
to strong clinical responses [1]. Since then, many other
targeted therapies have proved effective as long as the
cancer cells depend on the targeted oncogene. Examples
include antagonist antibodies against HER2 or VEGFR,
tyrosine kinase inhibitors (TKIs) against EGFR or HER2
and inhibitors of the BRAF kinase. Such targeted therapies
have increased the life expectancy of patients suffering from
cancers such as lung cancer, breast cancer, and melanoma
[2], provided the tumor is clearly oncogene-addicted.
Despite these encouraging results, the therapeutic ef-
cacy of such inhibitors is limited by the systematic
appearance, after weeks or months of treatment, of acquired
resistances leading to treatment failure. Many studies have
aimed to identify resistance mechanisms in cell models or
by genome resequencing of the relapse tumors. Because
most targeted therapies are directed against RTKs and sig-
naling kinases, the rst main mechanism of resistance
involves activation of alternative signaling pathways that
bypass driver inhibition. An example of this resistance
mechanism, called off-target resistance, is the case of lung
tumors harboring an activating EGFR mutation and treated
with an EGFR TKI: 1020% of patients treated with a rst-
generation EGFR TKI display EGFR-inhibition-bypassing
*David Tulasne
david.tulasne@ibl.cnrs.fr
1CNRS, Inserm, Institut Pasteur de Lille, CHU Lille, UMR9020-
U1277CANTHERCancer Heterogeneity Plasticity and
Resistance to Therapies, Univ. Lille, Lille, France
2Thoracic Oncology Department, CHU, Univ. Lille, Lille, France
Supplementary information The online version contains
supplementary material available at https://doi.org/10.1038/s41388-
021-01835-0.
1234567890();,:
1234567890();,:
MET gene amplication [3]. The second main mechanism
of resistance described is modication of the target itself by
additional molecular alterations which abolish the efcacy
of a targeted therapy. This resistance mechanism, called on-
target resistance, is also found in lung tumors harboring an
activating EGFR mutation: in about 50% of patients treated
with a rst-generation EGFR TKI, there appears an addi-
tional EGFR mutation at residue T790, which leads to
resistance by interfering with TKI binding [4].
In recent years, the receptor tyrosine kinase MET has
emerged as an important actor in resistance to various tar-
geted therapies. In this review, we describe how the MET
receptor triggers resistance of various cancers to therapies
targeting RTKs such as EGFR, VEGFR, and HER2 or
signaling hubs such as BRAF. In physiological situations,
interestingly, functional relationships have been described
between MET and the bypassed target, suggesting that
resistance stems from preexisting mechanisms. We also
describe the recently observed on-target resistance of lung
tumors harboring activating MET mutations.
Presentation of the MET receptor
MET is an RTK produced predominantly in cells of epi-
thelial origin. It is activated by its stromal ligand, hepato-
cyte growth factor (HGF), also called scatter factor [5].
HGF and MET are essential to embryonic development,
since knockout of either one affects formation of the epi-
thelial organs, placenta, muscles, and neurons [69]. In
adults, the HGF-MET pair is involved in physiological
processes such as epidermal healing and liver regeneration
[1013].
The extracellular part of MET contains an N-terminal
SEMA domain encompassing the αsubunit and the rst amino
acids of the βsubunit, followed by a plexinsemaphorin
integrin domain and four immunoglobulin-like domains. The
intracellular region contains a juxtamembrane domain, the
tyrosine kinase domain, and a C-terminal tail [14]. MET
interaction with its ligand favors its dimerization and autop-
hosphorylation of two tyrosine residues in the catalytic domain
(Y1234/Y1235). Other tyrosine residues are then phosphory-
lated, especially tyrosine 1003 in the juxtamembrane domain
and tyrosines 1349/1356 in the C-terminal tail [15,16]. The
phosphorylated C-terminal residues allow recruitment of
important effectors such as GAB1 and PI3K. This multi-
substrate docking site plays a key role in MET-induced bio-
logical responses, since its mutation in mice causes phenotypes
similar to those of MET-decient mice [17]. Downregulation
of MET receptor activity is also an essential process, as it
prevents receptor oversignaling. All the negative regulatory
mechanisms evidenced so far target the juxtamembrane region
ofthecytoplasmicpart,encodedbyexon14[18].
Resistance to EGFR TKIs: the prototype of
resistance involving MET
In non-small cell lung cancer (NSCLC), mutations affecting
the kinase domain of EGFR (epidermal growth factor
receptor) and inducing constitutive activation of this
receptor are found in 1040% of cases. MET and EGFR
have been shown to activate similar downstream pathways,
notably the MAPK and PI3K pathways [19]. Thus, inhibi-
tion of one of these two receptors can be compensated by
activation of the other. Direct crosstalk between MET and
EGFR has also been evidenced. It was rst shown that
EGFR ligands such as TGFalpha and EGF activate MET
through EGFRMET heterodimerization and that EGFR
and MET co-immunoprecipitate in tumor cells over-
expressing EGFR [20,21]. Ligand-independent activation
of MET by EGFR has also been demonstrated, in work
highlighting the role of G-protein coupled receptors in MET
transactivation in human carcinoma cells [22]. Inversely,
HGF has been found to induce transcription of Heparin-
Binding Epidermal Growth Factor (HB-EGF), a ligand of
EGFR [23]. MET can also activate EGFR indirectly,
through activation of SRC, which in turn activates EGFR
[24] (Fig. 1A). In addition, MET and HER-family members
often appear to be co-expressed in tumors [25,26]. EGFR
variants such as vIII EGFR have been found to induce MET
activation in glioblastomas [27].
The involvement of MET in resistance to EGFR-targeted
therapies was rst observed in EGFR-mutated NSCLC. In
this type of cancer, MET activation through MET gene
amplication acts as a bypass pathway leading to resistance
to EGFR TKIs [3]: MET amplication causes HER3
transactivation, which in turn induces activation of the PI3K
pathway. In this setting, cell proliferation can be abolished
only when an EGFR TKI and a MET TKI are combined.
Importantly, although the resistance due to MET ampli-
cation was rst observed in preclinical models, it has now
been conrmed by molecular analysis of paired tumor
samples obtained before and after exposure to an EGFR
TKI. MET amplication is found in 522% of samples
showing acquired resistance to a rst- or second-generation
EGFR TKI [3,28,29] (Fig. 1B). Variations in MET
amplication rates in the various series of post-progression
tumor samples may be due, in part, to the lack of a con-
sensual denition and technique for detection of MET
amplication [30]. Interestingly, clones displaying MET
amplication can be detected, at a very low rate, before any
treatment with an EGFR TKI, suggesting that these clones
are selected under therapeutic pressure [31]. Additionally,
MET amplication can occasionally but rarely occur toge-
ther with another mechanism of resistance to EGFR TKIs,
such as the EGFR T790M mutation [32]. Importantly, MET
amplication is also a mechanism of resistance to
4062 M. Fernandes et al.
Resistance mechanismsPhysiological interplay
P
Main treatment strategies
(Trial number or acronym)
MET
VEGFR
PTP1B
P
PTP1B
Hypoxia
P
bevacizumab
suninib
MET
HER2 HGF
trastuzumab
MET
expression
MET amplificaon
HGF expression
MET overexpression
HIF1a Signaling
Signaling
HCC, RCC
Breast cancer
CBA
MET EGFR Bispecific Anbodies MET/EGFR
-EMB-01 (NCT03797391)
-amivantanab (MARIPOSA/ PAPILLON)
-golvanib (NCT01332266)
Co-treatments MET TKI/EGFR TKI
-teponib/gefinib (INSIGHT)
-savolinib/osimernib (SAVANNAH/ ORCHARD)
-vannib/erlonib (MARQUEE)
Co-treatments MET TKI/ EGFR Ab
-capmanib/cetu ximab (NCT02205398)
-vannib/cetuximab (NCT01892527)
Co-treatments MET Ab/EGFR TKI
-emibetuzumab/erlonib (Chime)
Co-treatments HGF Ab/EGFR Ab
- rilotumumab/pan itumumab (NCT00788957)
Multarget TKI (MET/VEGFR/…)
-cabozannib (METEOR, MegaMOST, ACTION…)
-kaninib (NCT04093466)
-forenib (NCT00725764)
Co-treatments MET TKI/VEGFR TKI
-crizonib/axi nib (NCT01999972)
Co-treatments MET TKI/VEGFR Ab
-merisnib/ramucir umab (NCT02745769)
Co-treatments MET Ab/ VEGFR TKI
-onartuzumab/soraf enib (NCT01897038)
Co-treatments MET Ab/ VEGF Ab
- onartuzumab/bevacizumab (NCT01496742)
Co-treatment MET TKI/panHER TKI
-crizonib/dacominib (NCT01121575,
NCT01441128)
Co-treatments MET TKI/HER2 Ab
-cabozannib/trastu zumab (NCT02260531)
SRC P
HB-EGF
Dimerizaon
EGF
TGFα
or
HGF
HGF
cetuximab
panitumumab
MET amplificaon
Signaling
NSCLC HGF expression
1st generaon
erlonib
3rd generaon
osimernib
(T790M inhibion)
P
PP
PPP
BRAF PP2A P
MET amplificaon
PP2A
P
BRAF
BRAFi
HGF
HGF expression
PSer985
P
Signaling
MET
BRAF Melanoma
Co-treatement MET TKI/BRAF inhibitor
-crizonib/vemurafenib (NCT01531361)
-cabozannib/v emurafenib (NCT01835184)
-golvanib/sora fenib (NCT01271504)
Ser985
PPP
Fig. 1 Resistance mechanisms involving the MET receptor and
main therapeutic strategies. Illustrations summarizing examples of
physiological interplay (A), the corresponding resistance mechanisms
(B), and the main treatment strategies evaluated in clinical trials for
countering resistances due to EGFR/MET, VEGFR/MET, HER2/
MET, and BRAF/MET interplay (C). Hepatocellular carcinoma HCC,
Renal cell carcinoma RCC.
When the MET receptor kicks in to resist targeted therapies 4063
osimertinib, a third-generation EGFR TKI designed to tar-
get even T790M-bearing EGFR. This is true both when
osimertinib is used after failure of a rst EGFR TKI [33,34]
and when it is used as rst-line treatment [35] (Fig. 1B). In
colorectal cancer, MET amplication is responsible for
primary and acquired resistance to monoclonal antibodies
against EGFR. In patients whose tumors initially respond to
cetuximab or panitumumab and later progress without any
evidence of a KRAS mutation, MET amplication may be a
frequent mechanism of resistance [36] (Fig. 1B).
In addition to conferring resistance to EGFR TKIs, MET
amplication promotes an aggressive phenotype in EGFR-
mutated cells: increased cell proliferation, anchorage-
independent growth, and migration, leading to an
increased capacity to metastasize [37]. Patients displaying
MET amplication as a mechanism of resistance to EGFR
TKIs are more prone to develop new metastases and to have
a shorter time on treatment [37,38].
Beside to MET amplication, in both EGFR-mutated and
EGFR-wild-type cell lines, the MET ligand HGF has been
also shown to induce resistance to EGFR inhibitors,
including anti-EGFR antibodies and EGFR TKIs [39,40]
(Fig. 1B). On a larger scale, HGF can promote resistance to
several targeted therapies in models of oncogene addiction,
including EGFR- and BRAF-mutated cells [41]. In vitro,
HGF causes activation of the PI3K-AKT pathway via
GAB1, without recruitment of HER3 [31,39]. Transient
exposure to HGF is sufcient to promote clonal expansion
of MET-amplied clones among EGFR-mutated cells [31].
In a study where tumor samples were obtained from patients
with EGFR-mutated NSCLC after tumor progression on an
EGFR TKI, 61% of the samples displayed elevated levels of
HGF [42]. Patients showing tumor progression on an EGFR
TKI showed higher serum levels of HGF than patients with
no progression, and elevated serum HGF levels were sig-
nicantly associated with shorter progression-free and
overall survival [43]. HGF-induced resistance to EGFR
inhibitors could be overcome with mTOR inhibitors [44]or
by combining an EGFR TKI with either an antiangiogenic
agent [45] or a PI3K inhibitor [46].
Lastly, MET mutations have also been implicated in
acquired resistance to EGFR TKIs in EGFR-mutated
NSCLC. Suzawa et al. reported the case of an EGFR-
mutated NSCLC patient who, at the time of progression on
erlotinib, had developed both a T790M EGFR mutation and
a MET mutation leading to exon 14 skipping, and was later
stabilized with a combination of osimertinib and the MET
TKI crizotinib [47]. Furthermore, MET kinase mutations
have been proposed as a potential mechanism of acquired
resistance to EGFR TKIs in EGFR-mutated NSCLC [48].
Clinical studies have evaluated the combined use of an
EGFR TKI and a MET TKI in patients with MET-induced
resistance to EGFR inhibitors, mostly in EGFR-mutated
NSCLC (Table 1and Fig. 1C). MET-dependent resistance
was variously dened in these different studies, according
to different criteria (gene copy number (GCN) gain or the
ratio of MET GCN to centromeres) and thresholds. Com-
bining osimertinib with the specic MET TKI savolitinib, in
patients with MET amplication dened as GCN 5or
ratio 2, yielded an objective response rate of 30% in those
who had been treated previously with osimertinib and
6467% in those who had been treated previously with a
rst- or second-generation EGFR TKI [49]. In a randomized
phase II study, a combination of getinib and tepotinib, a
MET-specic TKI, was evaluated in MET-altered, EGFR-
mutated NSCLC patients having acquired resistance to an
EGFR TKI. In patients with MET amplication (GCN 5or
ratio 2), the median OS was 37.3 months for patients
treated with getinib and tepotinib, as compared to
13.1 months in those treated with chemotherapy [50]. In
another study, evaluating the combination of getinib and
capmatinib in patients with tumors progressing on EGFR
TKIs, the authors found antitumor activity to depend on the
level of MET amplication [51]. Other agents such as
amivantamab, a bispecic monoclonal antibody targeting
both EGFR and MET, have shown encouraging results in
MET-deregulated NSCLC, in both in vivo and clinical
studies [52,53].
Resistance to antiangiogenic VEGF and
VEGFR inhibitors
Angiogenesis, which ensures oxygenation and nutrient
uptake, is required for tumor growth. Therefore, investiga-
tors have developed therapies targeting angiogenic factors
such as VEGFR 1 and its ligand the VEGF. Since 2005, the
FDA has approved these therapies for gastrointestinal
stromal tumors, advanced renal cell carcinoma (RCC),
thyroid carcinoma, hepatocellular carcinoma (HCC), and
lung cancer [54]. The agents used include TKIs against
VEGFR (e.g., sorafenib, sunitinib, pazopanib, and vande-
tanib) and monoclonal antibodies such as bevacizumab and
ramucirumab (directed against VEGF and VEGFR2,
respectively).
Unfortunately, resistances to these inhibitors also arise.
They seem due, in part, to adaptive responses induced by
intratumoral hypoxia resulting from treatment with anti-
angiogenic compounds. MET receptor involvement in the
adaptive response was highlighted in the early 2000s:
hypoxia-induced factor 1 (HIF1a) promotes MET expres-
sion leading to increased survival, migration, and angio-
genesis [55]. Resistance to VEGF/VEGFR inhibitors is thus
proposed to involve the MET receptor as a consequence of
targeted-therapy-induced hypoxia. Furthermore, MET and
VEGFR2 have been found to interact directly. This
4064 M. Fernandes et al.
Table 1 Clinical trials evaluating multitarget agents or treatment combinations.
HGF/MET inhibitor combined with Target of TKI (inib) or Antibody (mab) Identier Acronym Phase Indication Ref.
VEGFR/MET
Cabozantinib MET/VEGFR2/Ret/Kit/Flt-1,3,4/Tie2/AXL TKI NCT03541902 CABOSUN II Metas. RCC [129]
NCT01755195 II Soft tissue sarcomas
NCT04134390 CABOMAYOR II Metas. RCC
NCT03425201 NICARAGUA I/II With Niraparib advanced UC
NCT04116541 MegaMOST II Malignant solid tumor
NCT04316182 ACTION II HCC
NCT01865747 METEOR III Metastatic RCC [68,130]
Kanitinib MET/VEGFR TKI NCT04093466 I Advanced solid tumor
Foretinib MET/VEGFR2/RON/AXL TKI NCT00725764 II Neoplasms, head and neck
Crizotinib Axitinib MET/ALK/ROS TKI +VEGFR TKI NCT01999972 Ib Advanced solid tumors [64]
Golvatinib Lenvatinib MET/VEGFR TKI +pan-VEGFR TKI NCT01433991 I/II Glioblastoma/melanoma
Merestinib Ramucirumab Multitarget MET TKI +anti-VEGFR2 NCT02745769 I Advanced cancers
Emibetuzumab Ramucirumab anti-MET +anti-VEGFR2 NCT02082210 I/II Advanced cancer
Onartuzumab Sorafenib anti-MET +VEGFR TKI NCT01897038 I Advanced HCC
Bevacizumab anti-MET +anti-VEGF NCT01496742 II NSCLC [131]
pan-HER
Crizotinib Dacomitinib MET/ALK/ROS TKI +pan-HER TKI NCT01121575 I NSCLC [80]
NCT01441128 I Advanced NSCLC
Foretinib Lapatinib MET/VEGFR2 TKI +pan-HER TKI NCT01138384 II Metas. Breast cancer [132]
HER2
Cabozantinib Trastuzumab MET/AXL/VEGFR2 TKI +anti-HER2 NCT02260531 II Breast cancer (brain Metas.) [133]
EGFR/MET
LY3164530 anti-MET/EGFR bispe.Ab NCT02221882 I Neoplasms/neoplasm Metas. [128]
EMB-01 anti-MET/EGFR bispe.Ab NCT03797391 I/II Advanced/Metas. Solid tumors
Emibetuzumab Erlotinib anti-MET +EGFR TKI NCT01900652 Chime II NSCLC
Savolitinib Osimertinib MET TKI +EGFR TKI NCT03778229 SAVANNAH II EGFRm+/MET+NSCLC after
Osimertinib
[134]
NCT03944772 ORCHARD II Advanced NSCLC after Osimertinib
Tivantinib Erlotinib MET TKI +EGFR TKI NCT01244191 MARQUEE III NSCLC [135]
Cetuximab MET TKI +anti-EGFR NCT01892527 II EGFR Resist/MET High; Metas. CC
Glumetinib Osimertinib MET TKI +EGFR TKI NCT04338243 I/II Metas. T790M negative and MET
amplif. NSCLC
Capmatinib Getinib MET TKI +EGFR TKI NCT01610336 II EGFR mut., MET amplif. NSCLC
after EGFRi
When the MET receptor kicks in to resist targeted therapies 4065
Table 1 (continued)
HGF/MET inhibitor combined with Target of TKI (inib) or Antibody (mab) Identier Acronym Phase Indication Ref.
Erlotinib MET TKI +EGFR TKI NCT02468661 I EGFR mutated, MET positive NSCLC
Nazartinib MET TKI +EGFR TKI NCT02335944 I/II EGFR-mutant NSCLC
NCT03333343 I EGFR-mutant NSCLC
Cetuximab MET TKI +anti-EGFR NCT02205398 I MET pos.; metas. RCC/HNSCC after
Cetuximab
EGFR/MET
Cabozantinib Erlotinib MET TKI +EGFR TKI NCT03213626 II EGFR +MET Expressiong metas. PA
Tepotinib Getinib MET TKI +EGFR TKI NCT01982955 INSIGHT II NSCLC [50,136]
Osimertinib MET TKI +EGFR TKI NCT03940703 INSIGHT 2 II Osimertinib relapsed MET amplif. NSCLC
Cetuximab MET TKI +anti-EGFR NCT04515394 II Colorectal neoplasms
Foretinib Erlotinib Multitarget MET TKI +EGFR TKI NCT01068587 I/II Advanced or metastatic NSCLC [137]
Onartuzumab Erlotinib anti-MET +EGFR TKI NCT01887886 III EGFR Mutated, MET pos. NSCLC [138]
Amivantamab Lazertinib anti-MET/EGFR bispe.Ab +EGFR TKI NCT04487080 MARIPOSA III EGFR-Mutated NSCLC
NCT02609776 CHRYSALIS I Advanced NSCLC
NCT04077463 I Advanced NSCLC
Carboplatin-
Pemetrexed
anti-MET/EGFR bispec.Ab +chemo NCT04538664 PAPILLON III Advanced or Metas. NSCLC with EGFR
Exon 20 Ins.
Golvatinib Cetuximab anti-MET +anti-EGFR NCT01332266 I/II SCC of the head and neck
Rilotumumab Panitumumab anti-HGF +anti-EGFR NCT00788957 I/II Metastatic CC [139]
Ficlatuzumab Getinib anti-HGF +EGFR TKI NCT01039948 I/II NSCLC [140]
Erlotinib anti-HGF +EGFR TKI NCT02318368 FOCAL II NSCLC
BRAF
Crizotinib Vemurafenib MET/ALK/ROS TKI +B-Raf inhibitor NCT01531361 I Advanced cancers with BRAF mut
Cabozantinib Vemurafenib Multitarget MET TKI +B-Raf inhib. NCT01835184 I Solid tumors or melanoma
Golvatinib Sorafenib MET/VEGFR TKI +Multitarget TKI Raf-1/B-
Raf/VEGFR-2, 3
NCT01271504 I/II HCC
Overview of the clinical trials registered at ClinicalTrial.gov (November 2020) evaluating the safety and/or efcacy of EGFR, HER2, VEGFR, or BRAF inhibitors (TKIs or antagonist monoclonal
antibodies) in combination with MET or HGF inhibitors (TKIs or antagonist monoclonal antibodies). For each clinical trial, the identier number, acronym, phase, main indications, and published
references are indicated.
RCC Renal cell carcinoma, HCC hepatocellular carcinoma, NSCLC non-small cell lung cancer, CC colorectal cancer, HNSCC head and neck squamous cell carcinoma, PA pancreatic
adenocarcinoma, SCC squamous cell carcinoma, UC urothelial cancer, Chemo. chemotherapy, Meta. metastatic or metastasis, amplif. amplication, Mut. mutated, progres. progression, Resist.
resistant, pos. positive.
4066 M. Fernandes et al.
interaction is involved in activated-VEGFR-mediated
downregulation of MET phosphorylation through
increased association of PTP1B phosphatase with MET.
Hence, VEGFR2 inhibition by the anti-VEGF bevacizumab
restores MET phosphorylation [56] (Fig. 1A). This nding
makes MET one of the main suspects in resistance to
VEGF/VEGFR inhibitors.
Meanwhile, many TKIs targeting two or more RTK-
family members have been developed. Cabozantinib,
among others, belongs to this multikinase inhibitor family.
Its targets notably include VEGFR and MET [57]. Such
inhibitors have been used in both preclinical models and
patients to evidence the involvement of MET in resistance
to VEGFR inhibition. They constitute promising tools for
treating resistant patients.
Especially for metastatic RCC, antiangiogenic therapy is
a major treatment modality. Unfortunately, about 20% of
patients are refractory to this approach, and a large majority
of remaining patients develop resistance [58]. Interestingly,
patient treatment with the VEGFR TKI sunitinib appears
associated with increased MET phosphorylation without
any concomitant increase in MET expression. In RCC cell
lines, chronic sunitinib treatment also leads to MET acti-
vation, associated with increased motility and invasion. In a
mouse model, both co-treatment of tumor xenografts with a
VEGFR-targeting and a MET-targeting TKI and treatment
with cabozantinib, which targets both, prevented growth of
sunitinib-resistant tumors, conrming the involvement of
MET activation in resistance [59,60] (Fig. 1B).
In HCC, the VEGFR inhibitor sorafenib, which also
inhibits several targets, has been approved for rst-line
treatment [61] (Table 1). For patients, however, the survival
benet of sorafenib treatment is limited. Preclinical testing
in animal models has revealed that the initial decrease in
angiogenesis under VEGFR inhibition is followed by rapid
revascularization, associated with a more invasive and
metastatic behavior [62]. Similarly, although mice xeno-
grafted with HCC cells show inhibition of tumor growth
when treated with a VEGF antagonist antibody, this effect is
concomitant with increased metastasis [63]. It is associated
with an increase in both HIF1a and MET expression, sug-
gesting that the hypoxia associated with VEGF-targeting
therapy promotes MET expression. The involvement of the
MET receptor in this resistance was evidenced by the ef-
cacy of co-treatment with an anti-VEGF Ab and a MET
TKI, which prevents both tumor growth and metastasis.
Interestingly, the multikinase inhibitor NZ001 under
development, which inhibits both VEGFR2 and MET,
induces a similar response [63]. Unfortunately, the results of
the rst study of combination treatment with axitinib (a
selective inhibitor of VEGFR 13) and crizotinib failed to
demonstrate a clinical benet for patients with advanced
RCC [64].
In NSCLC, bevacizumab is currently the only anti-
angiogenic agent approved for rst-line treatment. Yet the
clinical benets of these therapies for NSCLC are modest,
giving rise to both intrinsic and eventually acquired resis-
tances [65]. In preclinical models, xenograft tumors derived
from NSCLC cell lines showed, upon treatment with var-
ious VEGFR TKIs, acquired resistance associated with
increased HGF and MET expression leading to its phos-
phorylation. Ectopic expression of HGF in NSCLC cell
lines reproduced this resistance, and co-treatment with
VEGFR and MET TKIs overcame it, conrming the
potential role of HGF in causing resistance. Interestingly,
plasma analysis of NSCLC patients enrolled in clinical trials
evaluating the efcacy of the VEGFR TKI vandetanib
revealed an association between elevated circulating levels
of HGF and poor prognosis [66].
In neuroblastoma, likewise, MET activation by its ligand
may be a resistance mechanism, as the IGR-N91 neuro-
blastoma cell line, resistant to the VEGFR TKI axitinib,
displays elevated levels of HGF and of the phosphorylated
form of MET. In addition, treating the parental cell line with
HGF triggers resistance to axitinib. Consistently with these
observations, the growth of tumors derived from these
neuroblastoma cell lines was inhibited by co-treatment with
the VEGFR- and MET-targeting TKI cabozantinib, this
effect being associated with decreased angiogenesis and
increased apoptosis [67].
Following the positive results from the phase 3
METEOR trial [68] (Table 1and Fig. 1C), the multikinase
TKI cabozantinib was approved by the FDA in 2016, rst
for treatment of patients with advanced RCC having
received prior antiangiogenic therapy and for rst-line
treatment. It is unclear, however, whether the efcacy of
cabozantinib is due to its ability to prevent the appearance
of resistance mechanisms involving interplay between
VEGFR and MET or to other causes.
Resistance to anti-HER2 in breast cancer
The HER2 receptor is overexpressed in ~1530% of inva-
sive breast cancers and also in gastric and gastroesophageal
junction adenocarcinomas. This overexpression is associated
with reduced disease-free and overall survival. HER2+
breast tumors generally display constitutive tyrosine phos-
phorylation, signalling pathway activation, and a high rate of
cell growth [69].
The development of anti-HER2 agents such as mono-
clonal antibodies against HER2 (trastuzumab), TKIs (lapa-
tinib, afatinib, dacomitinib able to inhibit HER-family
RTKs), and an antibody-cytotoxic agent conjugate (trastu-
zumab emtasine) has dramatically improved breast cancer
patient care. Yet in the majority of patients, tumor
When the MET receptor kicks in to resist targeted therapies 4067
progression is observed in the year after initiation of tras-
tuzumab treatment [70]. Mechanisms of resistance to anti-
HER2 agents are wide-ranging. Resistance can be asso-
ciated with loss of HER2 through either decreased receptor
expression, masking of its extracellular domain or shedding
of the trastuzumab target domain through the action of
proteases [69]. Furthermore, one frequently observes
alterations in signalling pathways downstream of HER2,
such as activating mutations in PI3K/AKT/mTOR pathway
members [71] or SRC [72].
The involvement of the MET receptor in resistance to
anti-HER2 treatments was evidenced by transcriptomic
analysis of a HER2-positive breast cancer cohort (Fig. 1B).
Interestingly, tumors resistant to anti-HER2 treatments
showed higher MET expression [25]. MET amplication is
observed in approximately one-fourth of HER2+breast
cancer cases and is associated with a higher risk of trastu-
zumab therapy failure [73]. Similarly, HER2+gastric
cancer studies also revealed MET amplication as a
mechanism of resistance to the pan-HER inhibitor afatinib
[74,75]. Consistently with these clinical observations,
trastuzumab treatment of HER2-positive breast cancer cell
lines led to increased MET expression, involved in the
observed acquired resistance [25].
Interestingly, the interplay between HER2 and MET is
key to explaining how METparticularly its over-
expressionpromotes resistance to HER2 inhibition.
Indeed, trastuzumab efciently inhibits HER2 homo-
dimerization but not HER2 heterodimerization with other
RTKs. Yet, the MET receptor is able to dimerize with
HER2, and this interaction is not inhibited by trastuzumab.
Consequently, the HER2/MET interaction leads to cross-
activation of the two receptors and to induction of both
RTK downstream signaling pathways [76] (Fig. 1A). This
co-activation triggers synergistic action promoting cell
invasion in epithelial cell models [77].
HGF overexpression has also been detected in resistant
tumors [25] and can be associated with trastuzumab failure
[73]. In HER2+metastatic gastric cancers, Takashi et al.
evidenced a signicant association between a low serum
level of HGF at trastuzumab pretreatment and a positive
response, a high level being a factor of poor prognosis [78].
Furthermore, an increased serum level of HGF, as compared
to pretreatment, was observed in cases of trastuzumab
failure, suggesting that serum HGF could be used as a
tracker of the response to trastuzumab.
In the light of MET involvement in resistance to HER2
inhibition, strategies combining MET TKI treatment with
HER2 inhibition by an antibody or a TKI were rst eval-
uated in cell models. Shattuck et al. found MET inhibition
to restore sensitivity to trastuzumab treatment in HER2+
breast cancer cell lines [25]. Similarly, in HER2-amplied
gastric cancer cell lines rendered resistant to afatinib by
MET amplication, co-therapy against HER2 and MET was
found to inhibit tumor growth [75]. A combination of
lapatinib and foretinib (a multikinase inhibitor of MET and
VEGFR) was likewise found to reverse resistance to lapa-
tinib [79]. These data support the relevance of developing
anti-MET strategies to ght resistance to therapies targeting
HER2 (Table 1and Fig. 1C). Unfortunately, the phase I
study (NCT01121575) that investigated the effect of com-
bining dacomitinib, a pan-HER inhibitor, with crizotinib in
patients with advanced-NSCLC showed limited antitumor
activity and substantial toxicity [80].
Resistance to BRAF inhibitors in melanoma
and anaplastic thyroid carcinoma
More than half of metastatic melanomas display BRAF-
activating mutations, mainly BRAF-V600E, leading to
constitutive activation of the MEKERK signaling pathway
[81]. These patients can be treated with a combination
including a BRAF inhibitor (BRAFi) such as vemurafenib.
Such treatments have proved effective in about 50% of
patients, but various innate and acquired resistances can
lead to treatment failure [82,83]. Many resistance
mechanisms have been described, including reactivation of
the MAPK signaling pathway or upregulation of PI3K-
AKT-mTOR [84].
The involvement of HGF/MET signaling in resistance to
BRAFis was rst described in two back-to-back papers
published in Nature in 2012. The objective of both studies
was to investigate, in a large panel of cell lines with various
oncogene addictions, whether sensitivity to targeted thera-
pies could be overcome either by co-culturing with stromal
cells [85] or by adding various RTK ligands [41]. Both
studies revealed HGF as an effective inducer of resistance to
BRAFi in melanoma cell lines expressing the BRAF-
V600E oncogene (Fig. 1B), by preventing cell death and by
restoring activation of the MEKERK and PI3KAKT
signaling pathways. The MET TKI crizotinib was able to
prevent this response, demonstrating that HGF-induced
resistance was dependent on MET.
HGF expression analysis in a cohort of patients with
BRAF-V600E melanoma revealed its expression in the
tumor-associated stroma, with an increase of this expression
after treatment with BRAFi [85]. Also, an increase in circu-
lating HGF in BRAF-mutant melanoma patients was asso-
ciated with shorter progression-free and overall survival [41].
Besides resistances involving HGF expression, an acti-
vating BRAF mutation (G469A) was recently found to
coexist with MET gene amplication in a patient-derived
xenograft from a liver metastasis. Although MET ampli-
cation usually leads to constitutive MET activation, the
receptor was found, unexpectedly, to be inactivated. Its
4068 M. Fernandes et al.
activation was rescued upon BRAF inhibition. This sug-
gests an interplay between these two oncogenes [86], a view
supported mechanistically by the fact that BRAF signaling
inhibits the PP2A phosphatase known to dephosphorylate
Ser985 of MET. Since Ser985 phosphorylation inhibits
MET kinase activity, BRAF activation leads to inhibition of
MET activity, and the MET amplication remains func-
tionally dormant. Treatment with a BRAFi reactivates this
alteration, causing resistance to this targeted therapy (Fig.
1A). However, this elegant mechanism have to be con-
rmed in other patients. On the basis of all these data, phase
I clinical trials are ongoing to evaluate the use of MET TKIs
and BRAFis in advanced BRAF-mutated cancers, including
melanomas (Table 1and Fig. 1C).
In anaplastic thyroid carcinoma (ATC), BRAF-V600E is
again the most common oncogenic driver, often associated
with a TP53 alteration [87]. A recent clinical trial of
vemurafenib treatment for ATC showed signicant respon-
ses, but these were followed by resistance [88]. To investi-
gate the underlying resistance mechanisms, an in vivo mouse
model of ATC was developed, based on both thyroid-
specic TP53 inactivation and doxycycline-dependent
BRAF-V600E expression [89]. Dox withdrawal induced
complete tumor regression, followed by high-rate recurrence,
constituting an elegant model of resistance. Analysis of
tumors resistant to BRAF blockade revealed focal ampli-
cation of chromosome 6, causing both MET and HGF
overexpression. The involvement of MET activation in the
resistance process was then demonstrated in the model by
effective inhibition of tumor growth by MET inhibitors [89].
In thyroid cancer cell lines with BRAF-V600E mutation,
acquired resistance was likewise associated with MET
amplication leading to reactivation of the PI3K/AKT sig-
naling pathway [90]. Further study is required, however, to
conrm the existence of this resistance mechanism in patient.
In metastatic colorectal cancer, BRAF mutations are found
in about 10% of patients. Yet in a phase II clinical trial,
selective BRAFis such as vemurafenib showed only a
minimal effect against BRAF-mutant CRC [91]. This inef-
cacy as compared to the results obtained in melanoma
patients appeared associated with reactivation of EGFR sig-
naling [92]. This led to evaluating, in clinical trials, co-
treatment with BRAFi and EGFR-antagonist antibodies [93].
In the context of these co-treatments, novel mechanisms of
resistance were revealed, including amplication or mutation
of KRAS and also MET amplication. Pietrantonio et al. [94]
report that a patient co-treated with the anti-EGFR antibody
panitumumab and the BRAFi vemurafenib displayed disease
progression after an initial response. This resistance to tar-
geted co-treatment was associated with MET amplication
[36]. In the same study, interestingly, MET overexpression in
colorectal cancer cells was found to cause resistance to
BRAF and EGFR inhibition. Subsequent co-treatment of the
patient with vemurafenib and the MET TKI crizotinib proved
effective. This conrmed MET gene amplication as a
resistance mechanism in this context [94]. As previously
mentioned, this observation has to be conrmed in other
patients in order to assess how widespread this mechanism is.
On-target resistance in METexon14 NSCLC
patients
Although MET has been identied as a driver oncogene in
several well-characterized situations [95], it was not until
this year that the clinical use of MET-targeting therapies
was approved for treatment of NSCLC [96,97] (Supple-
mentary Table S1).
Novel MET-gene alterations, called METex14 mutations,
have recently been found in about 3% of lung tumors
[98,99]. All these alterations affect splice sites of MET exon
14 and lead to in-frame exon 14 skipping and subsequent
deletion of its regulatory juxtamembrane domain [18]. This
domain contains several negative regulatory sites, including
the ligand-dependent phosphorylated tyrosine 1003 and
serine 985. The Y1003 is involved in the recruitment of the
E3 ubiquitin ligase CBL involved in MET ubiquitination, a
molecular mechanism underlying intracellular RTK traf-
cking [100] and Ser985 phosphorylated by protein kinase
C is involved in feedback downregulation of the MET
kinase activity [101]. Juxtamembrane domain contains also a
caspase cleavage site (ESVD1002), leading upon apoptotic
stress to generation of a cytoplasmic fragment able to
amplify cell death through mitochondrial permeabilization
[102104].
Importantly, objective responses to several MET-
targeting TKIs have been observed in NSCLC patients
harboring METex14 mutations. This was the case, for
instance, in two phase II trials: VISION (evaluated tepoti-
nib) and GEOMETRY (evaluated capmatinib). In these
trials, about half of the METex14 patients showed an
objective response, and the median duration of the response
exceeded 11 months [97,105]. These results have led the
Japanese Ministry of Health to approve tepotinib for the
treatment advanced METex14 NSCLC. In the US, tepotinib
treatment has received breakthrough therapy designation
and capmatinib has been approved for patients with meta-
static METex14 NSCLC.
Thus, in the last few years, various MET TKIs (crizoti-
nib, glesatinib, capmatinib, tepotinib) have been used and
have shown efcacy in clinical trials or as off-label treat-
ments for METex14 NSCLC patients [96,97]. Yet after
initial responses and as expected for targeted therapies,
resistances have systematically been observed. Many
resistance mechanisms have been described, mostly invol-
ving activation of the usual suspects such as other RTKs or
When the MET receptor kicks in to resist targeted therapies 4069
downstream signaling hubs [106,107]. Some resistances,
however, are due to on-target mutations, essentially in the
MET kinase domain. In the context of this review we can
thus state that MET resists its own targeting.
To date, seven mutations have been clinically described,
after treatment with a MET TKI, in patients harboring
METex14 mutations: H1067, H1112, G1181, L1213,
F1218, D1246, and Y1248 [108118]. These mutations
usually emerge after only a few months of treatment with
crizotinib, glesatinib, or capmatinib (Table 2and Fig. 2A).
The presence of multiple mutations in the kinase domain
has also been observed [108,109,114,116,117]. For
example, D1246N/H has been found with Y1248H [109]
and G1181R can be present with L1213V, D1246H/N, and
Y1248H/S [116] in the patients circulating DNA.
In the recent study of Recondo et al. among 20
METex14-mutation-harboring patients who experienced
resistance to a MET TKI, 7 (35%) displayed on-target
resistances due to MET mutations, the other resistances
being off-target ones [116].
Most of the MET mutations recently found to be asso-
ciated with resistance to TKIs were already known as MET-
activating mutations, notably in renal cancer [119122].
Detection of these mutations in tumors suggests that they
may play a direct role in resistance through ligand-
independent MET activation.
Resistance to MET TKIs has also been investigated in
cell models expressing METex14 and/or displaying
METex14 oncogene dependence. In Ba/F3 cells engi-
neered to depend, for their growth, on ectopic expression
of an oncogene rather than on a cytokine, 12 MET resis-
tance mutations were revealed, including ve at residues
previously spotted in patients (G1181, L1213, F1218,
D1246, and T1248) [123]. Interestingly, mutations at
positions 1246 and 1248 were involved in resistance to
type I TKIs, which bind the active form of MET, while
mutations at 1213 and 1218 were involved in resistance to
type II TKIs, which bind the inactive form. This study thus
suggests that the contribution of specic mutations to
resistance may depend on the type of TKI [123]. These
Table 2 Acquired MET
mutations after MET TKI in
METex14 NSCLC, review of
clinical data.
Ref. Sample type before
resistance
TKI Sample site at
resistance
MET mutation post TKI
[109] cfDNA Crizotinib cfDNA D1246N; D1246H; Y1248H
[112] Tumor Crizotinib Metastasis Liver D1246N
[118] Tumor Crizotinib cfDNA G1181R; D1246H;
D1246A; Y1248H
[110] Tumor Crizotinib Tumor +cfDNA D1246N, Y1248H/S, G1181R,
(+glenatinib L1213V)
[113] Tumor +cfDNA Crizotinib cfDNA D1246N
[108]Crizotinib D1246N
[108]Crizotinib Tumor +cfDNA Y1248H; D1246N; D1246S
Glesatinib cfDNA D1246N; G1181R; L1213V
Glesatinib Tumor +cfDNA H1067Y; L1213V; L1213F
Crizotinib cfDNA D1246N
[117] cfDNA Crizotinib cfDNA Y1248S; F1218I
cfDNA Crizotinib cfDNA Y1248H; D1248N
cfDNA Crizotinib cfDNA L1213V
cfDNA Crizotinib cfDNA D1246H
[116] Tumor biopsy Glesatinib Tumor +cfDNA H1112Y; L1213V
Tumor biopsy Crizotinib Tumor +cfDNA G1181R; L1213V; D1246H/N;
Y1248H/S
Tumor biopsy Crizotinib Tumor D1246H
Tumor biopsy Crizotinib cfDNA D1246N
Tumor biopsy Capmatinib Tumor +cfDNA D1246N
Tumor biopsy Crizotinib Tumor +cfDNA Y1248C
Tumor biopsy Glesatinib-
Crizotinib
Tumor MET Amplication
This table compiles the data available from recent publications describing MET kinase mutations identied
in METex14 patients treated with the indicated MET TKI. Site and type of the collected samples before and
after resistance are indicated.
cfDNA Circulating free DNA.
4070 M. Fernandes et al.
data are also in agreement with the view that a con-
formational change induced by a particular mutation in the
kinase domain may interfere with binding of particular
MET TKIs. For instance, the resistance mutations at
positions 1181 and 1248 are consistent with steric hin-
drance of crizotinib binding (crizotinib being a type I
TKI), while the resistance mutations at positions 1213 and
1218 are consistent with steric hindrance of binding of
merestinib, a type II TKI (Fig. 2B). Interestingly, MET
variants with resistance mutations against type I TKIs were
found to be sensitive to type II inhibitors, and vice versa
[123]. This suggests that resistance might be overcome
through the complementary activity of the MET TKIs.
Similar results were obtained in NIH3T3 cells, where
expression of a MET variant displaying both an exon 14
deletion and a D1246N or Y1248C mutation was found to
drive resistance to type I but not type II inhibitors [110]. In
the same publication, the authors report a patient with a
METex14mutationwhodisplayedresistancetocrizotinib,
associated with the presence of the following additional
MET mutations: D1246N, Y1248H/S, and G1181R. On
the basis of results obtained in cell models, treatment was
switched from the type I inhibitor crizotinib to the type II
inhibitor glesatinib. This led to regression of some meta-
static localizations, with disappearance of the Y1248H/S
mutation. This disappearance was concomitant, however,
with the appearance of an L1213V mutation [110], iden-
tied in a study by Fujino et al. [123] as a mutation leading
to resistance to type II TKIs.
On-target resistance has also been described in the con-
text of MET gene amplication. A case report describes a
patient with an initial EGFR-activating mutation who
METex14
MET TKI
crizonib
glesanib
capmanib
H1067Y
H1112Y
G1181R
L1213V/F
F1218I
D1246H/N/A/S
Y1248H/N/S/C
Acve MET kinase
with crizonib type I inhibitor
Inacve MET kinase
with meresnib type II inhibitor
A
CB
MET WT
Y1248
DFG
in conformaon
crizonib
G1181
DFG
out conformaon F1218
meresnib
L1213
Fig. 2 On-target resistance to
MET TKIs in NSCLC
displaying MET exon
14 skipping. A Illustration of
MET WT and MET exon 14,
with the positions of the on-
target resistance mutations that
appear within the kinase domain
upon treatment with MET TKIs.
B3D structure images of the
active form of MET kinase in a
complex with the type I inhibitor
crizotinib (DOI: 10.2210/
pdb2WGJ/pdb). C3D structure
images of the inactive form in a
complex with the type II
inhibitor merestinib (DOI:
10.2210/pdb4EEV/pdb). The
images were created with Mol*,
an online software of the
Research Collaboratory for
Structural Bioinformatics
Protein Data Bank (www.rcsb.
org)[141]. The position of the
DFG motif of the ATP pocket in
the in or out conformation is
colored in yellow, residue
G1181 in blue, Y1248 in red,
L1213 in pink, and F1218
in green.
When the MET receptor kicks in to resist targeted therapies 4071
developed primary resistance to an EGFR TKI through
MET amplication [124]. Co-treatment with an EGFR TKI
and the MET TKI savolitinib led to a dramatic clinical
response. After several months of co-treatment, however,
the patient experienced a relapse associated with detection
of an additional MET D1246V mutation, previously
described as an activating mutation [125]. In the same
study, interestingly, the authors demonstrated with recon-
stituted MET mutations in cell lines that the D1246V
mutant is resistant to type I TKIs, including savolitinib, but
not to type II MET inhibitors. This nding led to adapting
the treatment to an EGFR TKI and cabozantinib, a type II
MET TKI, to which the progressive lung disease responded
dramatically [124].
Taken together, case reports and cell-based resistance
assays allow proposing a owchart for sequential treatment
of METex14 lung cancer patients. Mutations causing
resistance to type I TKIs, including at least the D1246H/N
and Y1248H/N/S/C mutations, can be overcome with type
II MET TKIs. Conversely, mutations causing to type II
TKIs, including at least the L1213F and F1218I mutations,
can be overcome with type I TKIs (Fig. 3).
Conclusion and prospects
A review of the literature identies MET activation as an
effective mechanism of resistance to targeted therapies against
several RTKs including EGFR, HER2, and VEGFR (Fig. 4).
Inthesethreecases,theresistancecanbequalied as a sub-
stitution resistance, since MET can replace another RTK. One
would think that the substitution worksbecause of major
similarities between the downstream signaling pathways of
MET and of the replaced RTK. Yet when MET replaces
EGFR in the context of resistance to an EGFR TKI, only a
subset of signaling pathways seems involved in the resistance.
First, in cell models of oncogene addiction, specic targeting
of either MET or EGFR with a TKI led to similarly altered
phosphoprotein proles and to largely common transcriptomic
modications. Reductions in the phosphoproteome were
mainly dependent on RAS and PI3K, suggesting that the MET
and EGFR oncogene addictions involve similar signaling hubs
restricted to a few pathways [126]. The central role of RAS
and PI3K signaling in resistance was latter demonstrated by
the fact that mutations in and amplication of KRAS and PI3K
are widespread mechanisms of resistance to both EGFR and
MET TKIs [127]. Therefore, at least for EGFR/MET sub-
stitution, the resistance seems supported by restricted common
downstream signaling pathways including the RAS and PI3K
pathways.
Resistance through substitution also seems supported by
preexisting functional relationships between RTKs, includ-
ing their heterodimerization, cross-activation, and negative
feedback loops. This is exemplied by the interplay
between VEGFR and MET. The MET receptor participates
in the adaptive response to hypoxia, since MET, like
VEGFR, is an angiogenic receptor. Thus, hypoxia due to
VEGFR inhibition leads to MET re-expression, which in
turn supports resistance [55]. Furthermore, as activated-
VEGFR negatively regulates MET phosphorylation through
the phosphatase PTP1B, MET is directly activated upon
VEGFR inhibition [56]. Thus, resistance mechanisms
appear to rely both on similarities in RTK signaling and on
preexisting functional relationships between RTKs. These
observations suggest that acquired resistance may arise not
only through random phenomena but also through oriented
processes. First, resistance can emerge through selection of
the molecular alteration that is most appropriate for effec-
tive substitution. Second, resistance can be initiated by
breaking of a negative feedback loop by the targeted ther-
apy, leading to activation of the resistance factor.
The second main mechanism of resistance involving
MET is exemplied by resistance to BRAFis in melanoma
and thyroid carcinoma (Fig. 4). In these cancers, MET
activation through gene amplication can bypass inhibition
of BRAF. This mechanism is thus called bypass resistance.
Interestingly, as in the case of substitution resistance, the
existence of a functional relationship between BRAF and
MET may favor establishment of the resistance: BRAF-
inhibitor-triggered disruption of the negative feedback
exerted by BRAF on MET through PP2A promotes MET
activation.
METex14
MET amplif.
Type I TKI
Type II TKI
Type II TKI
METex14 +
D1246H/N
First line TKI On-target
Resistance
Second line TKIInial MET
alteraon
METex14 +
Y1248H/N/S/C
Type I TKI
METex14 +
L1213F
METex14 +
F1218I
crizonib
capmanib
teponib
savolinib
cabozannib
meresnib
glesanib
Fig. 3 Flowchart for potential sequential treatments of METex14
lung cancer patients. Results of case reports and cell-based resistance
assays have led to proposing a owchart for sequential treatment of
METex14 lung cancer patients with either type I or type II MET TKIs.
4072 M. Fernandes et al.
For all substitution and bypass resistances, the existence
of powerful MET TKIs opens the way to potential co-
treatments. In resistant cell lines and experimental animal
tumorigenesis, co-targeting of MET and of a second
oncogenic RTK (VEGFR, EGFR, or HER2) has proven
effective. Multiple trials are in progress to investigate co-
targeting with two inhibitors or with multitarget agents, and
results are pending (Table 1). Interestingly, some clinical
trials are specically designed to evaluate co-treatment
efcacy in the expected resistance situation. For example,
clinical trials are evaluating EGFR/MET TKI co-treatment
as a way to counter resistance associated with MET gene
amplication. One should note, however, that co-treatment
studies considering molecular MET alterations as stratiers
remain the exception. It is also important to mention the
possibility of targeting two receptors with a single com-
pound such as cabozantinib, a TKI that inhibits both
VEGFR and MET, and the bispecic antibodies which
target both EGFR and MET [52,128].
The third mechanism of resistance involving MET is its
own molecular alteration as a consequence of targeted
therapy against MET (Fig. 4). Such on-target resistance has
been observed in the context of METex14 mutations in lung
cancer: resistance arises through additional MET mutations
within the kinase domain. In this situation, the mutation
interferes directly with TKI-MET binding. Because type I
and type II MET inhibitors bind, respectively, to either the
active or the inactive kinase domain, different MET muta-
tions occur according to the type of treatment. In several
patient cases, these observations have led to proposing a
sequence of treatments to overcome resistance [110,124].
These case reports open the way to possible adaptation of
MET-targeting therapies according to the type of on-target
resistance (Fig. 3).
Basic knowledge of RTKs, accumulated from their dis-
covery in the early 80s, has led to the development of
targeted therapies which have profoundly changed the
treatment of eligible patients. Much has notably been
learned about the complex mechanisms which underlie
RTK activation and downstream signaling. In parallel with
improvements in targeted therapy, the focus of scientic
research on RTKs in general and on the MET receptor in
particular has shifted gradually from basic studies to applied
research, on the false premise that RTK signaling is suf-
ciently well known, since RTK targeting is effective. Yet
the emergence of resistances has revealed how shaky that
premise is. It now appears that most resistances are based on
preexisting functional links between target and resistance
factor. Hence, extensive functional data are needed to fully
understand resistance mechanisms. This includesbut is
not limited tothe study of RTK co-receptors, of signaling
hubs shared by RTKs, and of negative feedback loops
between RTKs and actors of the signaling pathways. Con-
stant cross feeding between basic and clinical research will
thus be required to meet future challenges imposed by
resistance to targeted therapies.
Acknowledgements This work was supported by the CNRS, the
Institut Pasteur de Lille, and INSERM, and by grants from the
Cancéropôle Nord-Ouest, the Ligue Contre le Cancer, Comités
Nord et Aisne, the Agence nationale de la rechercheand the
Institut National du Cancer.
Compliance with ethical standards
Conict of interest The author declares no competing interests.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
Off Target Resistance
EGFR
(NSCLC)
HGF
MET
amplificaon /overexpression
Signaling
HGF expression
VEGFR
(HCC,RCC)
HER2
(breast cancer)
Ab
BRAF
BRAFi
METex14
(NSCLC)
On Target Resistance
L1213V/F
F1218I
D1246H/N/A/S
Y1248H/N/S/C
Type II
TKI
Type I
TKI
Signaling
«Bypas
« Substuon »
TKI
(Melanoma)
Fig. 4 Off- and on-target
resistances involving the MET
receptor. Illustrations
summarizing the main interplay
and resistance mechanisms
involving the MET receptor.
HCC hepatocellular carcinoma,
RCC renal cell carcinoma,
NSCLC non-small cell lung
cancer.
When the MET receptor kicks in to resist targeted therapies 4073
References
1. OBrien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M,
Cervantes F, et al. Imatinib compared with interferon and low-
dose cytarabine for newly diagnosed chronic-phase chronic
myeloid leukemia. N Engl J Med. 2003;348:9941004.
2. Giovannetti E, Rodriguez JA. Targeted therapies in cancer:
where are we going? Cancer Drug Resistance. 2018;1:826.
3. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park
JO, et al. MET amplication leads to getinib resistance in lung
cancer by activating ERBB3 signaling. Science. 2007;316:103943.
4. Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto
RA, Brannigan BW, et al. Irreversible inhibitors of the EGF
receptor may circumvent acquired resistance to getinib. Proc
Natl Acad Sci USA. 2005;102:766570.
5. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF.
Met, metastasis, motility and more. Nat Rev Mol Cell Biol.
2003;4:91525.
6. Bladt F, Riethmacher D, Isenmann S, Aguzzi A, Birchmeier C.
Essential role for the c-met receptor in the migration of myogenic
precursor cells into the limb bud. Nature. 1995;376:76871.
7. Maina F, Hilton MC, Ponzetto C, Davies AM, Klein R. Met
receptor signaling is required for sensory nerve development and
HGF promotes axonal growth and survival of sensory neurons.
Genes Dev. 1997;11:334150.
8. Schmidt C, Bladt F, Goedecke S, Brinkmann V, Zschiesche W,
Sharpe M, et al. Scatter factor/hepatocyte growth factor is
essential for liver development. Nature. 1995;373:699702.
9. Uehara Y, Minowa O, Mori C, Shlota K, Kuno J, Noda T, et al.
Placental defect and embryonic lethality in mice lacking hepa-
tocyte growth factor/scatter factor. Nature. 1995;373:7025.
10. Calvi C, Podowski M, Lopez-Mercado A, Metzger S, Misono K,
Malinina A, et al. Hepatocyte growth factor, a determinant of
airspace homeostasis in the murine lung. PLoS Genet. 2013;9:
e1003228.
11. Borowiak M, Garratt AN, Wustefeld T, Strehle M, Trautwein C,
Birchmeier C. Met provides essential signals for liver regenera-
tion. Proc Natl Acad Sci USA. 2004;101:1060813.
12. Chmielowiec J, Borowiak M, Morkel M, Stradal T, Munz B,
Werner S, et al. c-Met is essential for wound healing in the skin.
J Cell Biol. 2007;177:15162.
13. Huh CG, Factor VM, Sanchez A, Uchida K, Conner EA,
Thorgeirsson SS. Hepatocyte growth factor/c-met signaling
pathway is required for efcient liver regeneration and repair.
Proc Natl Acad Sci USA. 2004;101:447782.
14. Schiering N, Knapp S, Marconi M, Flocco MM, Cui J, Perego
R, et al. Crystal structure of the tyrosine kinase domain of the
hepatocyte growth factor receptor c-Met and its complex with
the microbial alkaloid K-252a. Proc Natl Acad Sci USA. 2003;
100:126549.
15. Longati P, Bardelli A, Ponzetto C, Naldini L, Comoglio PM.
Tyrosines1234-1235 are critical for activation of the tyrosine
kinase encoded by the MET proto-oncogene (HGF receptor).
Oncogene. 1994;9:4957.
16. Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Gior-
dano S, et al. A multifunctional docking site mediates signaling
and transformation by the hepatocyte growth factor/scatter factor
receptor family. Cell. 1994;77:26171.
17. Maina F, Casagranda F, Audero E, Simeone A, Comoglio PM,
Klein R, et al. Uncoupling of Grb2 from the Met receptor
in vivo reveals complex roles in muscle development. Cell.
1996;87:53142.
18. Cortot AB, Kherrouche Z, Descarpentries C, Wislez M, Baldacci
S, Furlan A, et al. Exon 14 deleted MET receptor as a new bio-
marker and target in cancers. J Natl Cancer Inst. 2017;109:112.
19. Wagner JP, Wolf-Yadlin A, Sevecka M, Grenier JK, Root DE,
Lauffenburger DA, et al. Receptor tyrosine kinases fall into
distinct classes based on their inferred signaling networks. Sci
Signal. 2013;6:ra58.
20. Jo M, Stolz DB, Esplen JE, Dorko K, Michalopoulos GK, Strom
SC. Cross-talk between epidermal growth factor receptor and c-
Met signal pathways in transformed cells. J Biol Chem.
2000;275:880611.
21. Werbin JL, Avendaño MS, Becker V, Jungmann R, Yin P,
Danuser G, et al. Multiplexed Exchange-PAINT imaging reveals
ligand-dependent EGFR and Met interactions in the plasma
membrane. Sci Rep. 2017;7:12150.
22. Fischer OM, Giordano S, Comoglio PM, Ullrich A. Reactive
oxygen species mediate Met receptor transactivation by G
protein-coupled receptors and the epidermal growth factor
receptor in human carcinoma cells. J Biol Chem.
2004;279:289708.
23. Reznik TE, Sang Y, Ma Y, Abounader R, Rosen EM, Xia S,
et al. Transcription-dependent epidermal growth factor receptor
activation by hepatocyte growth factor. Mol Cancer Res.
2008;6:13950.
24. Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src
cooperate to compensate for loss of epidermal growth factor
receptor kinase activity in breast cancer cells. Cancer Res.
2008;68:331422.
25. Shattuck DL, Miller JK, Carraway KL, Sweeney C. Met receptor
contributes to trastuzumab resistance of Her2-overexpressing
breast cancer cells. Cancer Res. 2008;68:14717.
26. Nakajima M, Sawada H, Yamada Y, Watanabe A, Tatsumi M,
Yamashita J, et al. The prognostic signicance of amplication
and overexpression of c-met and c-erb B-2 in human gastric
carcinomas. Cancer. 1999;85:1894902.
27. Huang PH, Mukasa A, Bonavia R, Flynn RA, Brewer ZE,
Cavenee WK, et al. Quantitative analysis of EGFRvIII cellular
signaling networks reveals a combinatorial therapeutic strategy
for glioblastoma. Proc Natl Acad Sci USA. 2007;104:1286772.
28. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S,
Turke AB, Fidias P, et al. Genotypic and histological evolution
of lung cancers acquiring resistance to EGFR inhibitors. Sci
Transl Med. 2011;3:75ra26.
29. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao
W, et al. Analysis of tumor specimens at the time of acquired
resistance to EGFR-TKI therapy in 155 patients with EGFR-
mutant lung cancers. Clin Cancer Res. 2013;19:22407.
30. Camidge DR, Davies KD. MET copy number as a secondary
driver of epidermal growth factor receptor tyrosine kinase inhi-
bitor resistance in EGFR-mutant non-small-cell lung cancer. J
Clin Oncol. 2019;37:8557.
31. Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D,
Lifshits E, et al. Preexistence and clonal selection of MET
amplication in EGFR mutant NSCLC. Cancer cell. 2010;
17:7788.
32. Baldacci S, Mazieres J, Tomasini P, Girard N, Guisier F,
Audigier-Valette C, et al. Outcome of EGFR-mutated NSCLC
patients with MET-driven resistance to EGFR tyrosine kinase
inhibitors. Oncotarget. 2017;8:10510314.
33. Oxnard GR, Hu Y, Mileham KF, Husain H, Costa DB, Tracy P,
et al. Assessment of Resistance Mechanisms and Clinical
Implications in Patients With EGFR T790M-Positive Lung
Cancer and Acquired Resistance to Osimertinib. JAMA Oncol.
2018;4:152734.
34. Le X, Puri S, Negrao MV, Nilsson MB, Robichaux J, Boyle T,
et al. Landscape of EGFR-dependent andindependent resis-
tance mechanisms to osimertinib and continuation therapy
beyond progression in EGFR-Mutant NSCLC. Clin Cancer Res.
2018;24:6195203.
4074 M. Fernandes et al.
35. Ramalingam SS, Cheng Y, Zhou C, Ohe Y, Imamura F, Cho BC,
et al. Mechanisms of acquired resistance to rst-line osimertinib:
preliminary data from the phase III FLAURA study. Ann Oncol.
2018;29:viii740.
36. Bardelli A, Corso S, Bertotti A, Hobor S, Valtorta E, Siravegna
G, et al. Amplication of the MET receptor drives resistance to
anti-EGFR therapies in colorectal cancer. Cancer Discov.
2013;3:65873.
37. Baldacci S, Kherrouche Z, Cockenpot V, Stoven L, Copin MC,
Werkmeister E, et al. MET amplication increases the metastatic
spread of EGFR-mutated NSCLC. Lung Cancer. 2018;125:5767.
38. Lai GGY, Lim TH, Lim J, Liew PJR, Kwang XL, Nahar R, et al.
Clonal MET amplication as a determinant of tyrosine kinase
inhibitor resistance in epidermal growth factor receptor-mutant
non-small-cell lung cancer. J Clin Oncol. 2019;37:87684.
39. Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura
T, et al. Hepatocyte growth factor induces getinib resistance of
lung adenocarcinoma with epidermal growth factor receptor-
activating mutations. Cancer Res. 2008;68:947987.
40. Yamada T, Takeuchi S, Kita K, Bando H, Nakamura T, Mat-
sumoto K, et al. Hepatocyte growth factor induces resistance to
anti-epidermal growth factor receptor antibody in lung cancer. J
Thorac Oncol. 2012;7:27280.
41. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E,
et al. Widespread potential for growth-factor-driven resistance to
anticancer kinase inhibitors. Nature. 2012;487:5059.
42. Yano S, Yamada T, Takeuchi S, Tachibana K, Minami Y,
Yatabe Y, et al. Hepatocyte growth factor expression in EGFR
mutant lung cancer with intrinsic and acquired resistance to
tyrosine kinase inhibitors in a Japanese cohort. J Thorac Oncol.
2011;6:20117.
43. Kasahara K, Arao T, Sakai K, Matsumoto K, Sakai A, Kimura H,
et al. Impact of serum hepatocyte growth factor on treatment
response to epidermal growth factor receptor tyrosine kinase
inhibitors in patients with non-small cell lung adenocarcinoma.
Clin Cancer Res. 2010;16:461624.
44. Ishikawa D, Takeuchi S, Nakagawa T, Sano T, Nakade J, Nanjo
S, et al. mTOR inhibitors control the growth of EGFR mutant
lung cancer even after acquiring resistance by HGF. PLoS ONE.
2013;8:e62104.
45. Takeuchi S, Wang W, Li Q, Yamada T, Kita K, Donev IS, et al.
Dual inhibition of Met kinase and angiogenesis to overcome
HGF-induced EGFR-TKI resistance in EGFR mutant lung can-
cer. Am J Pathol. 2012;181:103443.
46. Donev IS, Wang W, Yamada T, Li Q, Takeuchi S, Matsumoto
K, et al. Transient PI3K inhibition induces apoptosis and over-
comes HGF-mediated resistance to EGFR-TKIs in EGFR mutant
lung cancer. Clin Cancer Res. 2011;17:22609.
47. Suzawa K, Ofn M, Schoenfeld AJ, Plodkowski AJ, Odintsov I,
Lu D, et al. Acquired MET Exon 14 alteration drives secondary
resistance to epidermal growth factor receptor tyrosine kinase
inhibitor in EGFR-mutated lung cancer. JCO Precis Oncol.
2019;3. https://doi.org/10.1200/PO.19.00011.
48. Schoenfeld AJ, Chan JM, Kubota D, Sato H, Rizvi H, Danesh-
bod Y, et al. Tumor analyses reveal squamous transformation
and off-target alterations as early resistance mechanisms to rst-
line osimertinib in EGFR-mutant lung cancer. Clin Cancer Res.
2020;26:265463.
49. Sequist LV, Han J-Y, Ahn M-J, Cho BC, Yu H, Kim S-W, et al.
Osimertinib plus savolitinib in patients with EGFR mutation-
positive, MET-amplied, non-small-cell lung cancer after pro-
gression on EGFR tyrosine kinase inhibitors: interim results from
a multicentre, open-label, phase 1b study. Lancet Oncol.
2020;21:37386.
50. Wu Y-L, Cheng Y, Zhou J, Lu S, Zhang Y, Zhao J, et al.
Tepotinib plus getinib in patients with EGFR-mutant non-
small-cell lung cancer with MET overexpression or MET
amplication and acquired resistance to previous EGFR inhibitor
(INSIGHT study): an open-label, phase 1b/2, multicentre, ran-
domised trial. Lancet Respir Med. 2020;8:113243.
51. Wu Y-L, Zhang L, Kim D-W, Liu X, Lee DH, Yang JC-H, et al.
Phase Ib/II study of capmatinib (INC280) plus getinib after
failure of epidermal growth factor receptor (EGFR) inhibitor
therapy in patients with EGFR-mutated, MET factor-dysregulated
non-small-cell lung cancer. J Clin Oncol. 2018;36:31019.
52. Moores SL, Chiu ML, Bushey BS, Chevalier K, Luistro L, Dorn
K, et al. A Novel Bispecic Antibody Targeting EGFR and cMet
Is Effective against EGFR Inhibitor-Resistant Lung Tumors.
Cancer Res. 2016;76:394253.
53. Haura EB, Cho BC, Lee JS, Han J-Y, Lee KH, Sanborn RE,
et al. JNJ-61186372 (JNJ-372), an EGFR-cMet bispecic anti-
body, in EGFR-driven advanced non-small cell lung cancer
(NSCLC). JCO. 2019;37:9009.
54. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of
anti-tumour activity. Nat Rev Cancer. 2008;8:57991.
55. Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano
S, Comoglio PM. Hypoxia promotes invasive growth by tran-
scriptional activation of the met protooncogene. Cancer Cell.
2003;3:34761.
56. Lu KV, Chang JP, Parachoniak CA, Pandika MM, Aghi MK,
Meyronet D, et al. VEGF inhibits tumor cell invasion and
mesenchymal transition through a MET/VEGFR2 complex.
Cancer Cell. 2012;22:2135.
57. Sennino B, Ishiguro-Oonuma T, Wei Y, Naylor RM, Williamson
CW, Bhagwandin V, et al. Suppression of tumor invasion and
metastasis by concurrent inhibition of c-Met and VEGF signaling
in pancreatic neuroendocrine tumors. Cancer Discov.
2012;2:27087.
58. Grifoen AW, Mans LA, de Graaf AMA, Nowak-Sliwinska P,
de Hoog C, de Jong TAM, et al. Rapid angiogenesis onset after
discontinuation of sunitinib treatment of renal cell carcinoma
patients. Clin Cancer Res. 2012;18:396171.
59. Ciamporcero E, Miles KM, Adelaiye R, Ramakrishnan S, Shen
L, Ku S, et al. Combination strategy targeting VEGF and HGF/c-
met in human renal cell carcinoma models. Mol Cancer Ther.
2015;14:10110.
60. Zhou L, Liu XD, Sun M, Zhang X, German P, Bai S, et al.
Targeting MET and AXL overcomes resistance to sunitinib
therapy in renal cell carcinoma. Oncogene. 2016;35:268797.
61. Bruix J, Raoul JL, Sherman M, Mazzaferro V, Bolondi L, Craxi
A, et al. Efcacy and safety of sorafenib in patients with
advanced hepatocellular carcinoma: subanalyses of a phase III
trial. J Hepatol. 2012;57:8219.
62. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H,
Vinals F, et al. Antiangiogenic therapy elicits malignant pro-
gression of tumors to increased local invasion and distant
metastasis. Cancer Cell. 2009;15:22031.
63. Zhang Y, Gao X, Zhu Y, Kadel D, Sun H, Chen J, et al. The dual
blockade of MET and VEGFR2 signaling demonstrates pro-
nounced inhibition on tumor growth and metastasis of hepato-
cellular carcinoma. J Exp Clin Cancer Res. 2018;37:93.
64. Michaelson MD, Gupta S, Agarwal N, Szmulewitz R, Powles T,
Pili R, et al. A phase Ib study of axitinib in combination with
crizotinib in patients with metastatic renal cell cancer or other
advanced solid tumors. Oncologist. 2019;24:1151e817.
65. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati
A, et al. Paclitaxel-carboplatin alone or with bevacizumab for
non-small-cell lung cancer. N Engl J Med. 2006;355:254250.
66. Cascone T, Xu L, Lin HY, Liu W, Tran HT, Liu Y, et al. The
HGF/c-MET pathway is a driver and biomarker of VEGFR-
inhibitor resistance and vascular remodeling in non-small cell
lung cancer. Clin Cancer Res. 2017;23:5489501.
When the MET receptor kicks in to resist targeted therapies 4075
67. Daudigeos-Dubus E, Le Dret L, Bawa O, Opolon P, Vievard A,
Villa I, et al. Dual inhibition using cabozantinib overcomes HGF/
MET signaling mediated resistance to pan-VEGFR inhibition in
orthotopic and metastatic neuroblastoma tumors. Int J Oncol.
2017;50:20311.
68. Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring
PN, Rini BI, et al. Cabozantinib versus everolimus in advanced
renal cell carcinoma (METEOR): nal results from a rando-
mised, open-label, phase 3 trial. Lancet. 2016;17:91727.
69. Vernieri C, Milano M, Brambilla M, Mennitto A, Maggi C, Cona
MS, et al. Resistance mechanisms to anti-HER2 therapies in
HER2-positive breast cancer: Current knowledge, new research
directions and therapeutic perspectives. Crit Rev Oncol/Hematol.
2019;139:5366.
70. Nahta R, Esteva FJ. Trastuzumab: triumphs and tribulations.
Oncogene. 2007;26:363743.
71. Loibl S, Majewski I, Guarneri V, Nekljudova V, Holmes E, Bria
E, et al. PIK3CA mutations are associated with reduced patho-
logical complete response rates in primary HER2-positive breast
cancer: pooled analysis of 967 patients from ve prospective
trials investigating lapatinib and trastuzumab. Ann Oncol.
2016;27:151925.
72. Zhang S, Huang WC, Li P, Guo H, Poh SB, Brady SW, et al.
Combating trastuzumab resistance by targeting SRC, a common
node downstream of multiple resistance pathways. Nat Med.
2011;17:4619.
73. Minuti G, Cappuzzo F, Duchnowska R, Jassem J, Fabi A,
OBrien T, et al. Increased MET and HGF gene copy numbers
are associated with trastuzumab failure in HER2-positive meta-
static breast cancer. Br J Cancer. 2012;107:7939.
74. Sanchez-Vega F, Hechtman JF, Castel P, Ku GY, Tuvy Y, Won
H, et al. EGFR and MET amplications determine response to
HER2 inhibition in ERBB2-amplied esophagogastric cancer.
Cancer Discov. 2019;9:199209.
75. Yoshioka T, Shien K, Takeda T, Takahashi Y, Kurihara E, Ogoshi
Y, et al. Acquired resistance mechanisms to afatinib in HER2-
amplied gastric cancer cells. Cancer Sci. 2019;110:254957.
76. Agarwal S, Zerillo C, Kolmakova J, Christensen JG, Harris LN,
Rimm DL, et al. Association of constitutively activated hepato-
cyte growth factor receptor (Met) with resistance to a dual
EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J
Cancer. 2009;100:9419.
77. Khoury H, Naujokas MA, Zuo D, Sangwan V, Frigault MM,
Petkiewicz S, et al. HGF converts ErbB2/Neu epithelial mor-
phogenesis to cell invasion. Mol Biol Cell. 2005;16:55061.
78. Takahashi N, Furuta K, Taniguchi H, Sasaki Y, Shoji H, Honma
Y, et al. Serum level of hepatocyte growth factor is a novel
marker of predicting the outcome and resistance to the treatment
with trastuzumab in HER2-positive patients with metastatic
gastric cancer. Oncotarget. 2016;7:492538.
79. Hassan MS, Williams F, Awasthi N, Schwarz MA, Schwarz RE,
Li J, et al. Combination effect of lapatinib with foretinib in HER2
and MET co-activated experimental esophageal adenocarcinoma.
Sci Rep. 2019;9:17608.
80. Jänne PA, Shaw AT, Camidge DR, Giaccone G, Shreeve SM,
Tang Y, et al. Combined Pan-HER and ALK/ROS1/MET inhi-
bition with dacomitinib and crizotinib in advanced non-small cell
lung cancer: results of a phase I study. J Thorac Oncol. 2016;
11:73747.
81. Cancer Genome Atlas Network. Genomic classication of
cutaneous melanoma. Cell. 2015;161:168196.
82. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P,
Larkin J, et al. Improved survival with vemurafenib in melanoma
with BRAF V600E mutation. N Engl J Med. 2011;364:250716.
83. Rizos H, Menzies AM, Pupo GM, Carlino MS, Fung C, Hyman
J, et al. BRAF inhibitor resistance mechanisms in metastatic
melanoma: spectrum and clinical impact. Clin Cancer Res.
2014;20:196577.
84. Obaid NM, Bedard K, Huang WY. Strategies for overcoming
resistance in tumours harboring BRAF mutations. Int J Mol Sci.
2017;18:115.
85. Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR,
Du J, et al. Tumour micro-environment elicits innate resistance to
RAF inhibitors through HGF secretion. Nature. 2012;487:5004.
86. Virzi AR, Gentile A, Benvenuti S, Comoglio PM. Reviving
oncogenic addiction to MET bypassed by BRAF (G469A)
mutation. Proc Natl Acad Sci USA. 2018;115:1005863.
87. Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah
RH, et al. Genomic and transcriptomic hallmarks of poorly dif-
ferentiated and anaplastic thyroid cancers. J Clin Investig.
2016;126:105266.
88. Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY,
et al. Vemurafenib in Multiple nonmelanoma cancers with
BRAF V600 mutations. N Engl J Med. 2015;373:72636.
89. Knauf JA, Luckett KA, Chen K-Y, Voza F, Socci ND, Ghossein
R, et al. Hgf/Met activation mediates resistance to BRAF inhi-
bition in murine anaplastic thyroid cancers. J Clin Investig.
2018;128:408697.
90. Byeon HK, Na HJ, Yang YJ, Kwon HJ, Chang JW, Ban MJ,
et al. c-Met-mediated reactivation of PI3K/AKT signaling con-
tributes to insensitivity of BRAF(V600E) mutant thyroid cancer
to BRAF inhibition. Mol Carcinog. 2016;55:167887.
91. Kopetz S, Desai J, Chan E, Hecht JR, ODwyer PJ, Maru D,
et al. Phase II pilot study of vemurafenib in patients with
metastatic BRAF-mutated colorectal cancer. J Clin Oncol.
2015;33:40328.
92. Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill
AP, et al. EGFR-mediated re-activation of MAPK signaling
contributes to insensitivity of BRAF mutant colorectal cancers to
RAF inhibition with vemurafenib. Cancer Discov. 2012;
2:22735.
93. Yaeger R, Cercek A, OReilly EM, Reidy DL, Kemeny N,
Wolinsky T, et al. Pilot trial of combined BRAF and EGFR
inhibition in BRAF. Clin Cancer Res. 2015;21:131320.
94. Pietrantonio F, Oddo D, Gloghini A, Valtorta E, Berenato R,
Barault L, et al. MET-driven resistance to dual EGFR and BRAF
blockade may be overcome by switching from EGFR to MET
inhibition in BRAF-mutated colorectal cancer. Cancer Discov.
2016;6:96371.
95. Duplaquet L, Kherrouche Z, Baldacci S, Jamme P, Cortot AB,
Copin MC, et al. The multiple paths towards MET receptor
addiction in cancer. Oncogene. 2018;37:320015.
96. Drilon A, Clark JW, Weiss J, Ou S-HI, Camidge DR, Solomon
BJ, et al. Antitumor activity of crizotinib in lung cancers har-
boring a MET exon 14 alteration. Nat Med. 2020;26:4751.
97. Paik PK, Felip E, Veillon R, Sakai H, Cortot AB, Garassino MC,
et al. Tepotinib in non-small-cell lung cancer with MET Exon
14 skipping mutations. N Engl J Med. 2020;383:93143.
98. Descarpentries C, Lepretre F, Escande F, Kherrouche Z, Figeac
M, Sebda S, et al. Optimization of Routine Testing for MET
Exon 14 Splice Site Mutations in NSCLC Patients. J Thorac
Oncol. 2018;13:187383.
99. The cancer genome atlas research Network. Comprehensive
molecular proling of lung adenocarcinoma. Nature. 2014;
511:54350.
100. Peschard P, Fournier TM, Lamorte L, Naujokas MA, Band H,
Langdon WY, et al. Mutation of the c-Cbl TKB domain binding
site on the Met receptor tyrosine kinase converts it into a
transforming protein. Mol Cell. 2001;8:9951004.
101. Hashigasako A, Machide M, Nakamura T, Matsumoto K. Bi-
directional regulation of Ser-985 phosphorylation of c-met via
protein kinase C and protein phosphatase 2A involves c-Met
4076 M. Fernandes et al.
activation and cellular responsiveness to hepatocyte growth
factor. J Biol Chem. 2004;279:2644552.
102. Tulasne D, Deheuninck J, Lourenco FC, Lamballe F, Ji Z, Leroy
C, et al. Proapoptotic function of the MET tyrosine kinase receptor
through caspase cleavage. Mol Cell Biol. 2004;24:1032839.
103. Duplaquet L, Leroy C, Vinchent A, Paget S, Lefebvre J, Vanden
Abeele F, et al. Control of cell death/survival balance by the
MET dependence receptor. eLife. 2020;9:122.
104. Lefebvre J, Muharram G, Leroy C, Kherrouche Z, Montagne R,
Ichim G, et al. Caspase-generated fragment of the Met receptor
favors apoptosis via the intrinsic pathway independently of its
tyrosine kinase activity. Cell Death Dis. 2013;4:e871.
105. Wolf J, Seto T, Han J-Y, Reguart N, Garon EB, Groen HJM,
et al. Capmatinib in MET Exon 14-mutated or MET-amplied
non-small-cell lung cancer. N Engl J Med. 2020;383:94457.
106. Jamme P, Fernandes M, Copin M-C, Descarpentries C, Escande
F, Morabito A, et al. Alterations in the PI3K pathway drive
resistance to MET inhibitors in NSCLC harboring MET Exon
14. J Thorac Oncol. 2020;15:74151.
107. Suzawa K, Ofn M, Lu D, Kurzatkowski C, Vojnic M, Smith
RS, et al. Activation of KRAS mediates resistance to targeted
therapy in MET Exon 14-mutant non-small cell lung cancer. Clin
Cancer Res. 2019;25:124860.
108. Awad MM, Bahcall M, Sholl LM, Wilson FH, Paweletz C,
Capelletti M, et al. Mechanisms of acquired resistance to MET
tyrosine kinase inhibitors (TKIs) in MET exon 14 (METex14)
mutant non-small cell lung cancer (NSCLC). J Clin Oncol.
2018;36:9069.
109. Dong H-J, Li P, Wu C-L, Zhou X-Y, Lu H-J, Zhou T. Response
and acquired resistance to crizotinib in Chinese patients with
lung adenocarcinomas harboring MET Exon 14 splicing alter-
nations. Lung Cancer. 2016;102:11821.
110. Engstrom LD, Aranda R, Lee M, Tovar EA, Essenburg CJ, Madaj
Z, et al. Glesatinib exhibits antitumor activity in lung cancer
models and patients harboring MET Exon 14 mutations and
overcomes mutation-mediated resistance to type I MET inhibitors
in nonclinical models. Clin Cancer Res. 2017;23:666172.
111. Guo R, Ofn M, Brannon AR, Chow A, Delasos L, Somwar R,
et al. MET inhibitor resistance in patients with MET exon 14-
altered lung cancers. J Clin Oncol. 2019;37:9006.
112. Heist RS, Sequist LV, Borger D, Gainor JF, Arellano RS, Le LP,
et al. Acquired resistance to crizotinib in NSCLC with MET
Exon 14 skipping. J Thorac Oncol. 2016;11:12425.
113. Jin W, Shan B, Liu H, Zhou S, Li W, Pan J, et al. Acquired
mechanism of crizotinib resistance in NSCLC with MET Exon
14 skipping. J Thorac Oncol. 2019;14:e1379.
114. Li A-N, Yang J, Zhang X-C, Zhang Z, Su J, Gou L-Y, et al.
Acquired MET Y1248H and D1246N mutations mediate resis-
tance to MET inhibitors in non-small cell lung cancer. Clin
Cancer Res. 2017;23:492937.
115. Ou S-HI, Young L, Schrock AB, Johnson A, Klempner SJ, Zhu
VW, et al. Emergence of preexisting MET Y1230C mutation as a
resistance mechanism to crizotinib in NSCLC with MET Exon
14 skipping. J Thorac Oncol. 2017;12:13740.
116. Recondo G, Bahcall M, Spurr LF, Che J, Ricciuti B, Leonardi
GC, et al. Molecular mechanisms of acquired resistance to MET
tyrosine kinase inhibitors in patients with MET Exon 14-mutant
NSCLC. Clin Cancer Res. 2020;26:261525.
117. Rotow JK, Gui P, Wu W, Raymond VM, Lanman RB, Kaye FJ,
et al. Co-occurring genetic alterations in the RAS pathway pro-
mote resistance to MET inhibitor treatment in non-small cell
lung cancer with a MET exon 14 skipping mutation. Clin Cancer
Res. 2020;26:43949.
118. Zhang Y, Yin J, Peng F. Acquired resistance to crizotinib in
advanced lung adenocarcinoma with MET exon 14 skipping.
Lung Cancer. 2017;113:6971.
119. Jeffers M, Fiscella M, Webb CP, Anver M, Koochekpour S,
Vande, Woude GF. The mutationally activated Met receptor
mediates motility and metastasis. Proc Natl Acad Sci USA.
1998;95:1441722.
120. Jeffers MF, Vande Woude GF. Activating mutations in the Met
receptor overcome the requirement for autophosphorylation of tyr-
osines crucial for wild type signaling. Oncogene. 1999;18:51205.
121. Maritano D, Accornero P, Bonifaci N, Ponzetto C. Two muta-
tions affecting conserved residues in the Met receptor operate via
different mechanisms. Oncogene. 2000;19:135461.
122. Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P,
et al. Germline and somatic mutations in the tyrosine kinase
domain of the MET proto-oncogene in papillary renal carcino-
mas. Nat Genet. 1997;16:6873.
123. Fujino T, Kobayashi Y, Suda K, Koga T, Nishino M, Ohara S,
et al. Sensitivity and resistance of MET Exon 14 mutations in
lung cancer to eight MET tyrosine kinase inhibitors in vitro. J
Thorac Oncol. 2019;14:175365.
124. Bahcall M, Sim T, Paweletz CP, Patel JD, Alden RS, Kuang Y,
et al. Acquired METD1228V mutation and resistance to MET
inhibition in lung cancer. Cancer Discov. 2016;6:133441.
125. Jeffers M, Schmidt L, Nakaigawa N, Webb CP, Weirich G,
Kishida T, et al. Activating mutations for the met tyrosine kinase
receptor in human cancer. Proc Natl Acad Sci USA. 1997;
94:1144550.
126. Bertotti A, Burbridge MF, Gastaldi S, Galimi F, Torti D, Medico
E, et al. Only a subset of Met-activated pathways are required to
sustain oncogene addiction. Sci Signal. 2009;2:ra80.
127. Cortot AB, Janne PA. Molecular mechanisms of resistance in
epidermal growth factor receptor-mutant lung adenocarcinomas.
Eur Respir Rev. 2014;23:35666.
128. Patnaik A, Gordon M, Tsai F, Papadopoulos KP, Rasco D, Beeram
M, et al. A phase I study of LY3164530, a bispecic antibody
targeting MET and EGFR, in patients with advanced or metastatic
cancer. Cancer Chemother Pharmacol. 2018;82:40718.
129. Flaifel A, Xie W, Braun DA, Ficial M, Bakouny Z, Nassar AH,
et al. PD-L1 expression and clinical outcomes to cabozantinib,
everolimus, and sunitinib in patients with metastatic renal cell
carcinoma: analysis of the randomized clinical trials METEOR
and CABOSUN. Clin Cancer Res. 2019;25:60808.
130. Escudier B, Powles T, Motzer RJ, Olencki T, Arén Frontera O,
Oudard S, et al. Cabozantinib, a new standard of care for patients
with advanced renal cell carcinoma and bone metastases? Subgroup
analysis of the METEOR trial. J Clin Oncol. 2018;36:76572.
131. Wakelee H, Zvirbule Z, De Braud F, Kingsley CD, Mekhail T,
Lowe T, et al. Efcacy and safety of onartuzumab in combina-
tion with rst-line bevacizumab- or pemetrexed-based che-
motherapy regimens in advanced non-squamous non-small-cell
lung cancer. Clin Lung Cancer. 2017;18:509.
132. Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH,
et al. A phase-I study of lapatinib in combination with foretinib, a
c-MET, AXL and vascular endothelial growth factor receptor
inhibitor, in human epidermal growth factor receptor 2 (HER-2)-
positive metastatic breast cancer. Breast Cancer Res. 2017;19:54.
133. Leone JP, Duda DG, Hu J, Barry WT, Trippa L, Gerstner ER,
et al. A phase II study of cabozantinib alone or in combination
with trastuzumab in breast cancer patients with brain metastases.
Breast Cancer Res Treat. 2020;179:11323.
134. Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W,
Ferrari D, et al. Randomized phase II study of erlotinib plus
tivantinib versus erlotinib plus placebo in previously treated non-
small-cell lung cancer. J Clin Oncol. 2011;29:330715.
135. Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A,
Barlesi F, et al. Phase III multinational, randomized, double-
blind, placebo-controlled study of tivantinib (ARQ 197) plus
erlotinib versus erlotinib alone in previously treated patients with
When the MET receptor kicks in to resist targeted therapies 4077
locally advanced or metastatic nonsquamous non-small-cell lung
cancer. J Clin Oncol. 2015;33:266774.
136. Shitara K, Yamazaki K, Tsushima T, Naito T, Matsubara N,
Watanabe M, et al. Phase I trial of the MET inhibitor tepotinib in
Japanese patients with solid tumors. Jpn J Clin Oncol. 2020;
50:85966.
137. Leighl NB, Tsao M-S, Liu G, Tu D, Ho C, Shepherd FA, et al. A
phase I study of foretinib plus erlotinib in patients with pre-
viously treated advanced non-small cell lung cancer: Canadian
cancer trials group IND.196. Oncotarget. 2017;8:6965162.
138. Spigel DR, Edelman MJ, OByrne K, Paz-Ares L, Mocci S, Phan
S, et al. Results from the phase III randomized trial of onartu-
zumab plus erlotinib versus erlotinib in previously treated stage
IIIB or IV non-small-cell lung cancer: METLung. J Clin Oncol.
2017;35:41220.
139. Van Cutsem E, Eng C, Nowara E, Swieboda-Sadlej A, Tebbutt
NC, Mitchell E, et al. Randomized phase Ib/II trial of rilotu-
mumab or ganitumab with panitumumab versus panitumumab
alone in patients with wild-type KRAS metastatic colorectal
cancer. Clin Cancer Res. 2014;20:424050.
140. Tan E-H, Lim W-T, Ahn M-J, Ng Q-S, Ahn JS, Shao-Weng Tan
D, et al. Phase 1b trial of clatuzumab, a humanized hepatocyte
growth factor inhibitory monoclonal antibody, in combination
with getinib in Asian patients with NSCLC. Clin Pharmacol
Drug Dev. 2018;7:53242.
141. Sehnal D, Rose AS, Koča J, Burley SK, Velankar S. Mol*:
towards a common library and tools for web molecular graphics.
In: Proceedings of the workshop on molecular graphics and
visual analysis of molecular data. Goslar, DEU: Eurographics
Association; 2018. pp 2933.
4078 M. Fernandes et al.
... MET dysregulation is a well-known mechanism that contributes to drug resistance to targeted cancer therapies [153]. Met hyperactivation was reported to account for about 22% of acquired resistance cases following first-generation EGFR-targeted therapy [154,155]. ...
... MET dysregulation is a well-established oncogenic mechanism driving cancer development, and it also mediates drug resistance to classical chemotherapy, targeted therapy, and immunotherapy in the treatment of cancer. Accumulating preclinical evidence suggests that MET is a druggable target for the circumvention of drug resistance [153,179]. The combination of EGFR and Met inhibitors in clinical settings has been proposed to overcome drug resistance because MET amplification is common in EGFR-mutated NSCLC patients not responding to EGFR-targeted therapies [156,193,194]. ...
Article
Full-text available
The Met protein is a cell surface receptor tyrosine kinase predominantly expressed in epithelial cells. Aberrant regulation of MET is manifested by numerous mechanisms including amplification, mutations, deletion, fusion of the MET proto-oncogene, and protein overexpression. They represent the common causes of drug resistance to conventional and targeted chemotherapy in numerous cancer types. There is also accumulating evidence that MET/HGF signaling drives an immunosuppressive tumor microenvironment and dampens the efficacy of cancer immunotherapy. Substantial research effort has been invested in designing Met-targeting drugs with different mechanisms of action. In this review, we summarized the current preclinical and clinical research about the development of Met-targeting drugs for cancer therapeutics. Early attempts to evaluate Met-targeted therapies in clinical trials without selecting the appropriate patient population did not produce satisfactory outcomes. In the era of personalized medicine, cancer patients harboring MET exon 14 alterations or MET amplification have been found to respond well to Met-inhibitor therapy. The application of Met inhibitors to overcome drug resistance in cancer patients is discussed in this paper. Given that kinases play critical roles in cancer development, numerous kinase-mediated signaling pathways are attractive targets for cancer therapy. Existing kinase inhibitors have also been repurposed to new kinase targets or new indications in cancer. On the other hand, non-oncology drugs have also been repurposed for treating cancer through kinase inhibition as one of their reported anticancer mechanisms.
... Type I and II inhibitors are both ATP competitive inhibitors, whereas type III inhibitors like tivantinib are non-ATP competitive allosteric inhibitors 6 . Unfortunately, despite the promising initial efficacy of c-MET inhibitors in clinical treatment, the inevitable drug resistance remains a challenge in almost all patients 7 . Thus, novel therapeutic strategies targeting c-MET are urgently needed to treat cancers and overcome c-MET acquired resistance. ...
Article
Full-text available
Various c-mesenchymal-to-epithelial transition (c-MET) inhibitors are effective in the treatment of non-small cell lung cancer; however, the inevitable drug resistance remains a challenge, limiting their clinical efficacy. Therefore, novel strategies targeting c-MET are urgently required. Herein, through rational structure optimization, we obtained novel exceptionally potent and orally active c-MET proteolysis targeting chimeras (PROTACs) namely D10 and D15 based on thalidomide and tepotinib. D10 and D15 inhibited cell growth with low nanomolar IC50 values and achieved picomolar DC50 values and >99% of maximum degradation (Dmax) in EBC-1 and Hs746T cells. Mechanistically, D10 and D15 dramatically induced cell apoptosis, G1 cell cycle arrest and inhibited cell migration and invasion. Notably, intraperitoneal administration of D10 and D15 significantly inhibited tumor growth in the EBC-1 xenograft model and oral administration of D15 induced approximately complete tumor suppression in the Hs746T xenograft model with well-tolerated dose-schedules. Furthermore, D10 and D15 exerted significant anti-tumor effect in cells with c-METY1230H and c-METD1228N mutations, which are resistant to tepotinib in clinic. These findings demonstrated that D10 and D15 could serve as candidates for the treatment of tumors with MET alterations.
... Les mécanismes de résistance aux thérapies conventionnelles et ciblées sont dynamiques et séquentiels. Ils impliquent des modifications phénotypiques réversibles, tels que des mécanismes de sénescence transitoire [31], de reprogrammation métabolique [32], des modifications épigénétiques [33], ou encore une transition épithélio-mésenchymateuse [34] et/ou des changements mutationnels irréversibles [35]. Ces processus sont difficilement observables chez les patients. ...
... Les mécanismes de résistance aux thérapies conventionnelles et ciblées sont dynamiques et séquentiels. Ils impliquent des modifications phénotypiques réversibles, tels que des mécanismes de sénescence transitoire [31], de reprogrammation métabolique [32], des modifications épigénétiques [33], ou encore une transition épithélio-mésenchymateuse [34] et/ou des changements mutationnels irréversibles [35]. Ces processus sont difficilement observables chez les patients. ...
Article
Full-text available
The recent emergence of tumor organoid cultures, or tumoroids, has enriched the repertoire of preclinical models in oncology. These microtumors are obtained in vitro by including cells from patient tumor samples in an extracellular matrix and cultured in specific media. Very close to the tumor of origin, tumoroids can be amplified fairly rapidly from a small quantity of tissue, established with high success rate for most tumor types, easily genetically engineered, and stored in biobanks. Tumoroids thus offer numerous possibilities in terms of basic research, such as the study of carcinogenesis or mechanisms of chemoresistance, but also the identification of new targets and preclinical validation of new anti-cancer compounds or personalized medicine. Technological developments and enrichment of tumoroids with other cell types are currently ongoing to optimally exploit the full potential of these models.
Article
Full-text available
Pediatric cancer continues to be a global health issue needing the pursuit of innovative treatment options. One emerging area is gene therapy, holding the potential for dramatic improvements in pediatric cancer care. This review comprehensively overviews recent developments and challenges in pediatric cancer gene therapy. It starts by explaining the concepts underlying gene therapy, including gene delivery systems, gene editing techniques, and the logic of targeted therapies. It then goes through recent advances, highlighting meaningful progress in using both viral and non-viral vectors to deliver genes to cancer cells and specifically effectively, it also examines the development and use of gene-editing tools like CRISPR/Cas9, being used to modify cancer-linked genes and enhance treatment results. This review offers a complete look at the different gene treatments used for cancers in kids. That includes cell therapies, viruses that target cancer cells, and gene tweaks that change the immune response. It breaks down how they work, what clinical trials show and where they fall short. The authors really dive into the challenges of taking these therapies from animal studies to actual patients - things like making good lab models and getting government approval. Most importantly, the review talks about the huge strides made recently in gene therapy for young cancer patients. but it also admits there's still a long way to go in making treatments better and improving quality of life. and Mapping out the landscape like this hopefully moves things along so more children can survive cancer.
Preprint
Full-text available
MET is a receptor tyrosine kinase (RTK) responsible for initiating signaling pathways involved in development and wound repair. MET activation relies on ligand binding to the extracellular receptor, which prompts dimerization, intracellular phosphorylation, and recruitment of associated signaling proteins. Mutations, which are predominantly observed clinically in the intracellular juxtamembrane and kinase domains, can disrupt typical MET regulatory mechanisms. Understanding how juxtamembrane variants, such as exon 14 skipping (METΔEx14), and rare kinase domain mutations can increase signaling, often leading to cancer, remains a challenge. Here, we perform a parallel deep mutational scan (DMS) of MET intracellular kinase domain in two fusion protein backgrounds: wild type and METΔEx14. Our comparative approach has revealed a critical hydrophobic interaction between a juxtamembrane segment and the kinase ⍺C helix, pointing to differences in regulatory mechanisms between MET and other RTKs. Additionally, we have uncovered a β5 motif that acts as a structural pivot for kinase domain activation in MET and other TAM family of kinases. We also describe a number of previously unknown activating mutations, aiding the effort to annotate driver, passenger, and drug resistance mutations in the MET kinase domain.
Preprint
Full-text available
MET is a receptor tyrosine kinase (RTK) responsible for initiating signaling pathways involved in development and wound repair. MET activation relies on ligand binding to the extracellular receptor, which prompts dimerization, intracellular phosphorylation, and recruitment of associated signaling proteins. Mutations, which are predominantly observed clinically in the intracellular juxtamembrane and kinase domains, can disrupt typical MET regulatory mechanisms. Understanding how juxtamembrane variants, such as exon 14 skipping (METΔEx14), and rare kinase domain mutations can increase signaling, often leading to cancer, remains a challenge. Here, we perform a parallel deep mutational scan (DMS) of MET intracellular kinase domain in two fusion protein backgrounds: wild type and METΔEx14. Our comparative approach has revealed a critical hydrophobic interaction between a juxtamembrane segment and the kinase αC helix, pointing to differences in regulatory mechanisms between MET and other RTKs. Additionally, we have uncovered a β5 motif that acts as a structural pivot for kinase domain activation in MET and other TAM family of kinases. We also describe a number of previously unknown activating mutations, aiding the effort to annotate driver, passenger, and drug resistance mutations in the MET kinase domain.
Article
Savolitinib is an oral MET (hepatocyte growth factor receptor) tyrosine kinase inhibitor, with demonstrated preliminary efficacy in several cancer types. Previous pharmacokinetics assessments showed that savolitinib is rapidly absorbed but there are limited data on the absolute bioavailability and absorption, distribution, metabolism, and excretion (ADME) of savolitinib. This open‐label, two‐part, phase 1 clinical study (NCT04675021) used a radiolabeled micro‐tracer approach to evaluate absolute bioavailability and a traditional approach to determine the ADME of savolitinib in healthy male adult volunteers (N = 8). Pharmacokinetics, safety, and metabolic profiling and structural identification from plasma, urine, and fecal samples were also assessed. Volunteers received a single oral savolitinib 600‐mg dose followed by intravenous 100 μg of [14C]savolitinib in Part 1 and a single oral 300‐mg [14C]savolitinib dose (≤4.1 MBq [megabecquerel] [14C]) in Part 2. Following Part 1, absolute oral bioavailability was 69%, the median time of maximum observed concentration was 3.5 hours, and the mean terminal half‐life was 6.1 hours. Following Part 2, 94% of the radioactivity administered was recovered, with 56% and 38% in urine and feces, respectively. Exposure to savolitinib and metabolites M8, M44, M2, and M3 accounted for 22%, 36%, 13%, 7%, and 2%, respectively, of plasma total radioactivity. Approximately 3% of the dose was excreted as unchanged savolitinib in urine. Most savolitinib elimination occurred via metabolism by several different pathways. No new safety signals were observed. Our data show that the oral bioavailability of savolitinib is high and the majority of savolitinib elimination occurs via metabolism and is excreted in the urine.
Article
Full-text available
Objectives: Tepotinib (MSC2156119J) is an oral, potent and highly selective small molecule mesenchymal-epithelial transition factor (MET) inhibitor for which the recommended Phase II dose of 500 mg once daily has been defined, based on the first-in-man trial conducted in the USA and Europe. We carried out a multicenter Phase I trial with a classic `3 + 3' design to determine the recommended Phase II dose in Japanese patients with solid tumors (NCT01832506). Methods: Patients aged ≥20 years with advanced solid tumors (refractory to standard therapy or for whom no effective standard therapy was available) received tepotinib at 215, 300 or 500 mg once daily in a 21-day cycle. Occurrence of dose-limiting toxicities during cycle 1 was used to determine the maximum tolerated dose. Efficacy, safety and pharmacokinetics were also evaluated to support the dose assessment. Results: Twelve patients were treated. Tepotinib was generally well tolerated with no observed dose-limiting toxicities; treatment-related adverse events were mainly grades 1-2. The tolerability profile of tepotinib was similar to that observed in non-Japanese populations. Pharmacokinetics in Japanese and Western patients was comparable. One patient with gastric cancer and one patient with urachal cancer had stable disease of ≥12 weeks in duration. The observed safety profile and pharmacokinetics are comparable with those in patients from the USA and Europe, and the recommended Phase II dose of tepotinib in Japanese patients was confirmed as 500 mg once daily. Conclusions: These results, including initial signals of antitumor activity, support further development of tepotinib in Japanese patients with cancer.
Article
Full-text available
Control of cell death/survival balance is an important feature to maintain tissue homeostasis. Dependence receptors are able to induce either survival or cell death in presence or absence of their ligand, respectively. However, their precise mechanism of action and their physiological importance are still elusive for most of them including the MET receptor. We evidence that pro-apoptotic fragment generated by caspase cleavage of MET localizes to the mitochondria-associated membrane region. This fragment triggers a calcium transfer from endoplasmic reticulum to mitochondria, which is instrumental for the apoptotic action of the receptor. Knock-in mice bearing a mutation of MET caspase cleavage site highlighted that p40MET production is important for FAS-driven hepatocyte apoptosis, and demonstrate that MET acts as a dependence receptor in vivo. Our data shed light on new signaling mechanisms for dependence receptors’ control of cell survival/death balance, which may offer new clues for the pathophysiology of epithelial structures.
Article
Full-text available
Purpose: Molecular mechanisms of acquired resistance to MET TKIs are poorly understood. We aimed to characterize the genomic mechanisms of resistance to type I and type II MET TKIs and their impact on sequential MET TKI therapy outcomes in patients with metastatic MET exon 14 mutant NSCLC. Experimental design: Genomic alterations occurring at the time of progression on MET TKIs were studied using plasma and tissue next-generation sequencing (NGS). Results: A total of 20 patients had tissue or plasma available for analysis at the time of acquired resistance to a MET TKI. Genomic alterations known or suspected to be mechanisms of resistance were detected in 15 patients (75%). On-target acquired mechanisms of resistance, including single and polyclonal MET kinase domain mutations in codons H1094, G1163, L1195, D1228, Y1230, and high levels of amplification of the MET exon 14 mutant allele, were observed in 7 patients (35%). A number of off-target mechanisms of resistance were detected in 9 patients (45%), including KRAS mutations and amplifications in KRAS, EGFR, HER3, and BRAF; one case displayed both on- and off-target mechanisms of resistance. In two patients with on-target resistant mutations, switching between type I and type II MET TKIs resulted in second partial responses. Conclusions: On-target secondary mutations and activation of bypass signaling drive resistance to MET TKIs. A deeper understanding of these molecular mechanisms can support the development of sequential or combinatorial therapeutic strategies to overcome resistance.
Article
9006 Background: MET exon 14 alterations comprise a novel class of lung cancer drivers. MET tyrosine kinase inhibitors (TKIs) are active in patients with these cancers, but objective response rates (ORRs) are modest (~30%-40%). A subset of these cancers may harbor intrinsic resistance. Moreover, patients with initial benefit invariably develop acquired resistance. We set out to identify potential resistance mechanisms. Methods: We studied patients with stage IV MET exon 14-altered lung cancers who received a MET TKI. When feasible, tumor and/or plasma samples were collected, prioritizing paired pre- and post-TKI collection. Tumor samples underwent targeted mass spectrometry analysis (Nantomics) and DNA- (including MSK-IMPACT)/RNA-based (MSK-Fusion) next-generation sequencing (NGS). Plasma cfDNA underwent targeted NGS. ORR and progression-free survival (PFS) were assessed (RECIST v1.1). Results: 74 patients received a MET TKI (1 TKI n = 55; ≥2 TKIs n = 19). 91% received crizotinib as their 1 st TKI. Pre-TKI MET levels in tumor tissue (range 0-2120 amol/µg) were associated with outcomes: ORR 63% (n = 7/11) and median PFS 6.9 mos with detectable MET vs ORR 0% (n = 0/5) and median PFS 4.6 mos with undetectable MET (HR for PFS 0.3). Pre-TKI RAS pathway activation was associated with response: ORR 0% (n = 0/6) with KRAS/NF1/RASA1 mutation vs ORR 29% (n = 25/87) in others. Similar outcomes were observed with pre-TKI KRAS expression (n = 16, all with detectable KRAS levels): ORR 0% (n = 0/2) in KRAS ≥700 amol/µg vs ORR 50% (n = 7/14) < 700 amol/µg. Acquired resistance (Jackman criteria) was seen in 29 patients, 9 with paired pre-/post-treatment samples. On-target acquired resistance was found in 2/9 patients (22%): MET D1228N (n = 1), HGF amplification (n = 1). Potential off-target acquired resistance mechanisms were found in 5/9 pts (44%): KRAS G13V (n = 1), RASA1 S742* (n = 1), MDM2 amplification (n = 2), EGFR amplification (n = 1). Conclusions: Lack of MET expression or RAS pathway activation is associated with poor MET TKI outcomes in MET exon14-altered lung cancers. On-target acquired resistance is found in < 25% of patients; HGF amplification is a novel mechanism. Off-target intrinsic/acquired resistance may be mediated by RAS/MDM2/EGFR pathway activation.
Article
Background: Among patients with non-small-cell lung cancer (NSCLC), MET exon 14 skipping mutations occur in 3 to 4% and MET amplifications occur in 1 to 6%. Capmatinib, a selective inhibitor of the MET receptor, has shown activity in cancer models with various types of MET activation. Methods: We conducted a multiple-cohort, phase 2 study evaluating capmatinib in patients with MET-dysregulated advanced NSCLC. Patients were assigned to cohorts on the basis of previous lines of therapy and MET status (MET exon 14 skipping mutation or MET amplification according to gene copy number in tumor tissue). Patients received capmatinib (400-mg tablet) twice daily. The primary end point was overall response (complete or partial response), and the key secondary end point was response duration; both end points were assessed by an independent review committee whose members were unaware of the cohort assignments. Results: A total of 364 patients were assigned to the cohorts. Among patients with NSCLC with a MET exon 14 skipping mutation, overall response was observed in 41% (95% confidence interval [CI], 29 to 53) of 69 patients who had received one or two lines of therapy previously and in 68% (95% CI, 48 to 84) of 28 patients who had not received treatment previously; the median duration of response was 9.7 months (95% CI, 5.6 to 13.0) and 12.6 months (95% CI, 5.6 to could not be estimated), respectively. Limited efficacy was observed in previously treated patients with MET amplification who had a gene copy number of less than 10 (overall response in 7 to 12% of patients). Among patients with MET amplification and a gene copy number of 10 or higher, overall response was observed in 29% (95% CI, 19 to 41) of previously treated patients and in 40% (95% CI, 16 to 68) of those who had not received treatment previously. The most frequently reported adverse events were peripheral edema (in 51%) and nausea (in 45%); these events were mostly of grade 1 or 2. Conclusions: Capmatinib showed substantial antitumor activity in patients with advanced NSCLC with a MET exon 14 skipping mutation, particularly in those not treated previously. The efficacy in MET-amplified advanced NSCLC was higher in tumors with a high gene copy number than in those with a low gene copy number. Low-grade peripheral edema and nausea were the main toxic effects. (Funded by Novartis Pharmaceuticals; GEOMETRY mono-1 ClinicalTrials.gov number, NCT02414139.).
Article
Background We evaluated the efficacy and safety of tepotinib, a potent and highly selective oral MET inhibitor, plus gefitinib in patients with epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) with MET overexpression (immunohistochemistry [IHC]2+ or IHC3+) or MET amplification having acquired resistance to EGFR inhibition. Methods In this open-label, phase 1b/2, multicentre, randomised trial (the INSIGHT study), we enrolled adult patients (≥18 years) with advanced or metastatic NSCLC, and Eastern Cooperative Oncology Group performance status of 0 or 1, from academic medical centres and community clinics in six Asian countries. In phase 1b, patients received oral tepotinib 300 mg or 500 mg plus gefitinib 250 mg once daily. In phase 2, patients with EGFR-mutant, T790M-negative NSCLC MET overexpression or MET amplification were randomly assigned (initially in a 1:1 ratio and then 2:1 following a protocol amendment) to tepotinib plus gefitinib at the recommended phase 2 dose or to standard platinum doublet chemotherapy. Randomisation was done centrally via an interactive voice-response system. The primary endpoint was investigator-assessed progression-free survival (PFS). Secondary endpoints included overall survival (OS) and safety. Subgroup analyses were preplanned in patients with high MET overexpression (IHC3+) or MET amplification (mean gene copy number ≥5 or MET to centromere of chromosome 7 ratio ≥2). Efficacy and patient characteristics were assessed on an intention-to-treat basis and safety was assessed for all patients who received at least one dose of study medication. Low recruitment led to early termination of phase 2, so all analyses are considered to be exploratory. This study is registered with ClinicalTrials.gov, NCT01982955, and the European Union Drug Regulating Authorities Clinical Trials Database, Eudra-CT 2016-001604-28. Findings From Dec 23, 2013, to May 25, 2017, 18 patients were enrolled in phase 1b (n=6 in the 300 mg tepotinib group; n=12 in the 500 mg tepotinib group) and 55 patients in phase 2 (n=31 in the tepotinib plus gefitinib group; n=24 in the chemotherapy group). No dose-limiting toxicities were observed in phase 1b, so tepotinib 500 mg was used as the recommended phase 2 dose. In phase 2, survival outcomes were similar between groups: median PFS was 4·9 months in the tepotinib plus gefitinib group (90% CI 3·9–6·9) versus 4·4 months in the chemotherapy group (90% CI 4·2–6·8; hazard ratio [HR] 0·67, 90% CI 0·35–1·28). Median OS was 17·3 months in the tepotinib plus gefitinib group (12·1–37·3) versus 18·7 months in the chemotherapy group (15·9–20·7; HR 0·69, 0·34–1·41). PFS and OS were longer with tepotinib plus gefitinib than with chemotherapy in patients with high (IHC3+) MET overexpression (n=34; median PFS 8·3 months [4·1–16·6] vs 4·4 months [4·1–6·8]; HR 0·35, 0·17–0·74; median OS 37·3 months [90% CI 24·2–37·3] vs 17·9 months [12·0–20·7]; HR 0·33, 0·14–0·76) or MET amplification (n=19; median PFS 16·6 months [8·3–not estimable] vs 4·2 months [1·4–7·0]; HR 0·13, 0·04–0·43; median OS 37·3 months [90% CI not estimable] vs 13·1 months [3·25–not estimable]; HR 0·08, 0·01–0·51). The most frequent treatment-related grade 3 or worse adverse events were increased amylase (5 [16%] of 31 patients) and lipase (4 [13%]) concentrations in the tepotinib plus gefitinib group and anaemia (7 [30%] of 23 patients) and decreased neutrophil count (3 [13%]) in the chemotherapy group. Interpretation Despite early study termination, in a preplanned subgroup analysis, our findings suggest improved anti activity for tepotinib plus gefitinib compared with standard chemotherapy in patients with EGFR-mutant NSCLC and MET amplification, warranting further exploration. Funding Merck KGaA.
Article
Background: A splice-site mutation that results in a loss of transcription of exon 14 in the oncogenic driver MET occurs in 3 to 4% of patients with non-small-cell lung cancer (NSCLC). We evaluated the efficacy and safety of tepotinib, a highly selective MET inhibitor, in this patient population. Methods: In this open-label, phase 2 study, we administered tepotinib (at a dose of 500 mg) once daily in patients with advanced or metastatic NSCLC with a confirmed MET exon 14 skipping mutation. The primary end point was the objective response by independent review among patients who had undergone at least 9 months of follow-up. The response was also analyzed according to whether the presence of a MET exon 14 skipping mutation was detected on liquid biopsy or tissue biopsy. Results: As of January 1, 2020, a total of 152 patients had received tepotinib, and 99 patients had been followed for at least 9 months. The response rate by independent review was 46% (95% confidence interval [CI], 36 to 57), with a median duration of response of 11.1 months (95% CI, 7.2 to could not be estimated) in the combined-biopsy group. The response rate was 48% (95% CI, 36 to 61) among 66 patients in the liquid-biopsy group and 50% (95% CI, 37 to 63) among 60 patients in the tissue-biopsy group; 27 patients had positive results according to both methods. The investigator-assessed response rate was 56% (95% CI, 45 to 66) and was similar regardless of the previous therapy received for advanced or metastatic disease. Adverse events of grade 3 or higher that were considered by investigators to be related to tepotinib therapy were reported in 28% of the patients, including peripheral edema in 7%. Adverse events led to permanent discontinuation of tepotinib in 11% of the patients. A molecular response, as measured in circulating free DNA, was observed in 67% of the patients with matched liquid-biopsy samples at baseline and during treatment. Conclusions: Among patients with advanced NSCLC with a confirmed MET exon 14 skipping mutation, the use of tepotinib was associated with a partial response in approximately half the patients. Peripheral edema was the main toxic effect of grade 3 or higher. (Funded by Merck [Darmstadt, Germany]; VISION ClinicalTrials.gov number, NCT02864992.).
Article
9009 Background: JNJ-372 binds EGFR and cMet to block ligand binding, promote receptor degradation, and trigger antibody-dependent cellular cytotoxicity in models of EGFR-mutated (EGFRm) NSCLC. Here we describe the ongoing phase 1 safety, pharmacokinetics (PK), and activity of JNJ-372 in patients (pts) with NSCLC, including 3 rd generation tyrosine kinase inhibitor (3GTKI)-relapsed EGFRm NSCLC and EGFR Exon20ins disease. Methods: Pts received JNJ-372 (140–1400 mg) IV weekly for the first 28-day cycle and biweekly thereafter. 1050–1400 mg doses are being explored in dose expansion. Blood samples were collected for PK analyses. Efficacy by investigator per RECIST v1.1 in pts with EGFRm NSCLC treated at ≥700 mg is presented. Tumors were characterized by next-generation sequencing of circulating tumor (ct)DNA and/or tumor tissue. Results: As of 17 Jan 2019, 116 enrolled pts with NSCLC were treated. Median age was 63 years, 38% were male, 77% were Asian, and 97% had EGFR mutations. Mean duration of treatment was 3.8 months, longest exposure was 20 cycles. The PK data set included pts from Korea (77%) and the US (23%). At the 1050 mg dose, 72% of pts achieved average concentrations above the EC 90 based on preclinical models. Adverse events (AEs; ≥20%) were rash (59%), infusion related reaction (58%), paronychia (28%), and constipation (22%). Additional EGFR/cMet-related AEs include stomatitis (17%), pruritis (15%), peripheral edema (11%), and diarrhea (7%). Grade ≥3 AEs were reported in 34% (8% treatment-related) with dyspnea (6%) and pneumonia (3%) most frequently observed. Among response-evaluable pts, 25/88 (28%) achieved best timepoint response of partial response (PR). 10/47 pts with prior 3GTKI therapy had best timepoint response of PR (6 confirmed), including 4 with C797S, 1 with cMet amplification, and 5 without identifiable EGFR/cMet-dependent resistance. 6/20 pts with Exon20ins had best timepoint response of PR (3 confirmed). Conclusions: JNJ-372 has a manageable safety profile consistent with EGFR and cMet inhibition. Preliminary responses were achieved in 3GTKI-relapsed disease, including C797S and cMet amplification, and Exon20ins disease; enrollment in dose expansion is ongoing. Clinical trial information: NCT02609776.
Article
Introduction MET TKIs (tyrosine kinase inhibitors) have demonstrated efficacy against advanced NSCLC with mutations causing MET exon 14 skipping (METex14 mutations), but primary resistance seems frequent, as response rates are lower than for targeted TKIs of other oncogene-addicted NSCLC. Given the known interplay between MET and PI3K, we hypothesized that in METex14 NSCLC, PI3K-pathway alterations might contribute to primary resistance to MET TKIs. Methods We reviewed clinical data from 65 patients with METex14 NSCLC, assessing PI3K-pathway alterations by targeted NGS (mutations) and immunohistochemistry (loss of PTEN). Using a cell line derived from a patient with primary resistance to a MET TKI and cell lines harboring both a METex14 mutation and a PI3K-pathway alteration, we assessed sensitivity to MET TKIs used alone or with a PI3K inhibitor and investigated relevant signaling pathways. Results We found a PIK3CA mutation in 2/65 samples (3%) and loss of PTEN in 6/26 (23%). All three of the MET-TKI-treated patients with a PI3K pathway alteration had shown progressive disease at first assessment. Likewise, MET TKIs had no effect on the proliferation of METex14-mutated cell lines with a PI3K-pathway alteration, including the PTEN-lacking patient-derived cell line. Treatment combining a MET TKI with a PI3K inhibitor caused inhibition of both PI3K and MAPK signaling and restored sensitivity to MET TKIs. Conclusion PI3K-pathway alterations are common in METex14 NSCLC and may confer primary resistance to MET TKIs. In preclinical models, PI3K inhibition restores sensitivity to MET TKIs.
Article
Background: Preclinical data suggest that EGFR tyrosine kinase inhibitors (TKIs) plus MET TKIs are a possible treatment for EGFR mutation-positive lung cancers with MET-driven acquired resistance. Phase 1 safety data of savolitinib (also known as AZD6094, HMPL-504, volitinib), a potent, selective MET TKI, plus osimertinib, a third-generation EGFR TKI, have provided recommended doses for study. Here, we report the assessment of osimertinib plus savolitinib in two global expansion cohorts of the TATTON study. Methods: In this multi-arm, multicentre, open-label, phase 1b study, we enrolled adult patients (aged ≥18 years) with locally advanced or metastatic, MET-amplified, EGFR mutation-positive non-small-cell lung cancer, who had progressed on EGFR TKIs. We considered two expansion cohorts: parts B and D. Part B consisted of three cohorts of patients: those who had been previously treated with a third-generation EGFR TKI (B1) and those who had not been previously treated with a third-generation EGFR TKI who were either Thr790Met negative (B2) or Thr790Met positive (B3). In part B, patients received oral osimertinib 80 mg and savolitinib 600 mg daily; after a protocol amendment (March 12, 2018), patients who weighed no more than 55 kg received a 300 mg dose of savolitinib. Part D enrolled patients who had not previously received a third-generation EGFR TKI and were Thr790Met negative; these patients received osimertinib 80 mg plus savolitinib 300 mg. Primary endpoints were safety and tolerability, which were assessed in all dosed patients. Secondary endpoints included the proportion of patients who had an objective response per RECIST 1.1 and was assessed in all dosed patients and all patients with centrally confirmed MET amplification. Here, we present an interim analysis with data cutoff on March 29, 2019. This study is registered with ClinicalTrials.gov, NCT02143466. Findings: Between May 26, 2015, and Feb 14, 2019, we enrolled 144 patients into part B and 42 patients into part D. In part B, 138 patients received osimertinib plus savolitinib 600 mg (n=130) or 300 mg (n=8). In part D, 42 patients received osimertinib plus savolitinib 300 mg. 79 (57%) of 138 patients in part B and 16 (38%) of 42 patients in part D had adverse events of grade 3 or worse. 115 (83%) patients in part B and 25 (60%) patients in part D had adverse events possibly related to savolitinib and serious adverse events were reported in 62 (45%) patients in part B and 11 (26%) patients in part D; two adverse events leading to death (acute renal failure and death, cause unknown) were possibly related to treatment in part B. Objective partial responses were observed in 66 (48%; 95% CI 39-56) patients in part B and 23 (64%; 46-79) in part D. Interpretation: The combination of osimertinib and savolitinib has acceptable risk-benefit profile and encouraging antitumour activity in patients with MET-amplified, EGFR mutation-positive, advanced NSCLC, who had disease progression on a previous EGFR TKI. This combination might be a potential treatment option for patients with MET-driven resistance to EGFR TKIs. Funding: AstraZeneca.