Summary:

The first KRASG12D inhibitor, MRTX113, leads to regression in multiple mouse models of PDAC as a monotherapy. MRTX113 blocks cancer cell proliferation, induces cancer cell death, and promotes immune infiltration and activation.

See related article by Kemp et al., p. 298 (6).

Pancreatic ductal adenocarcinoma (PDAC) has the highest death rate of the most commonly diagnosed cancers, and with a rising occurrence, it is predicted to become the second leading cause of cancer deaths by 2030 (1). One of the major determinants of the dire outcome of PDAC is the lack of effective targeted therapy. Dubbed the “undruggable” for several decades, mutational activation of the Kirsten rat sarcoma (KRAS) gene occurs in 95% of PDAC and has been unequivocally established as the initiating mutation and genetic driver of this disease (1). The KRAS protein is a GTPase that in response to cellular stimuli cycles between GDP-bound (OFF) and GTP-bound (ON) states to transmit intracellular signals. Under physiologic conditions, this system is controlled by guanine nucleotide exchange factors, which mediate the exchange of GDP for GTP, and GTPase accelerating proteins that accelerate the intrinsic hydrolysis of GTP-bound RAS by several orders of magnitude. Oncogenic mutations of KRAS frequently involve single amino acid substitutions in residues G12, G13, and Q61, which hinder the capacity to hydrolyze GTP, thus favoring the GTP-bound (ON) state of the molecule and uncontrolled signaling. The prevalence and tissue distribution of KRAS mutation subtypes vary greatly. Overall, mutations in KRASG12 (D/C/V/R/A/) are the most prominent in cancer (89%), with G12D as the most frequent (36%), followed by G12V (23%) and G12C (14%). The most prevalent KRAS mutations in PDAC are G12D (∼41%) and G12V (∼34%), whereas G12R is less frequent (∼16%; ref. 1). Pioneering work by Shokat and colleagues in 2013 fundamentally changed the 4-decades-long view of KRAS as undruggable and paved the way for the development of the KRASG12C inhibitors AMG510 (sotorasib) and MRTX849 (adagrasib; refs. 2–4). These molecules form irreversible covalent bonds with the C12 residue of GDP-bound KRAS and have shown remarkable clinical outcomes in KRASG12C non–small cell lung cancer and colorectal cancer (3, 4). Recently, Mirati Therapeutics reported MRTX133, a reversible KRASG12D inhibitor with single-digit nanomolar IC50 for KRASG12D (5). MRTX133 binds both the GDP and GTP states of KRASG12D, locking the protein in its inactive form or providing steric hindrance to block effector binding in the active setting. Building on these prior findings, Kemp and colleagues set out to discover the long-awaited answer to the question of the efficacy of small-molecule inhibition of KRASG12D in the treatment of PDAC (6).

The authors used several immunocompetent mouse models of PDAC that recapitulate human disease, including syngeneic orthotopic implantations of clonal cell lines derived from the LSL-KRasG12D/+;LSLTrp53R172H/+;Pdx-1-Cre (KPC) model, the autochthonous KPC/Y (KPC mice expressing a YFP epithelial lineage tag) model, and subcutaneous implantations of clonal cell lines derived KPC/Y tumors (7). Within a short time frame (7 days), treatment with MRTX113 as a monotherapy resulted in unprecedented regressions in all models. Although highly efficacious throughout, the different models revealed variations in the response to MRTX113. As such, treatment of the orthotopic or autochthonous KPC/Y models resulted in substantial regression in 100% of the mice, whereas only ∼50% showed regression in the subcutaneous implantation model after 1 week of treatment. Differences in regression were not due to differential sensitivity of cell lines to MRTX113. In addition, MRTX113 was selective to KRASG12D, as it had no impact on RAS downstream signaling and growth of subcutaneous tumors from a syngeneic derivative of a KPC cell line in which D12 had been mutated to a cysteine (C). These outcomes are complemented by another recent study showing that 2 weeks of treatment with MRTX113 induced significant regression (30% or greater) in 11 of 25 KRASG12D tumors in xenograft models spanning five tissue types (8). The greatest impact, however, was observed in pancreatic cancer models in which MRTX113 resulted in significant regression in 73% of the mice, supporting the reported critical role of mutant KRAS in PDAC as the earliest oncogenic mutation and bona fide genetic driver. These studies provide compelling evidence for the potential clinical use of MRTX113 in KRASG12D-driven PDAC.

From a cancer cell–intrinsic perspective, MRTX113 appears to operate similarly to the KRASG12C inhibitors. As with KRASG12C inhibitors, MRTX113 treatment led to dose-dependent decreases in phospho-ERK1/2 levels, but not phospho-AKT or phospho-S6 levels, thus identifying the MAPK pathway as a dominant mediator of KRAS-driven tumorigenesis (Fig. 1). Kemp and colleagues identified strong inhibition of phospho-ERK1/2 in the first 2 days of treatment, accompanied by an increase in the apoptotic marker cleaved caspase-3 (CC3) and a decrease in the proliferation marker Ki-67 (6). The antitumor response of MRTX113, therefore, appears to be driven by both the induction of apoptosis and inhibition of growth. After 1 week of treatment, the inhibition of proliferation and phospho-ERK1/2 was maintained; however, the increase in CC3 was lost. The reduction in cell death over time hints at the reactivation of apoptotic-suppressive pathways. On this note, the KRASG12C inhibitors tell a tale of several intrinsic and acquired resistance mechanisms, including redundancy in parallel signaling pathways, upregulation of the EGFR pathway, and novel mutations in MYC, BRAF, and the KRAS protein itself. Although MRTX113 led to robust tumor regression, examples of resistance to MRTX113 were observed. Within this framework, the durability of the response of KRASG12D-driven PDAC to MRTX113 is an important consideration for patient applicability. The antitumor effect of MRTX113 shows significant durability in the short term, as 100% of mice in the subcutaneous model that reached complete regressions (CR) at 2 weeks of MRTX113 treatment remained disease free for 3 subsequent weeks without therapy. KPC/Y mice with deep regression also remained in remission up to 2 weeks after therapy, but subsequently, tumors progressed in both models. Therefore, a thorough evaluation of biochemical events following KRASG12D inhibition is imperative for the identification of mechanisms of intrinsic and emerging resistance and optimal combinatorial therapies that circumvent them. These downstream changes in signaling should also be evaluated in a temporal manner, as it may be critical to pair certain therapies at different time points of treatment. Interestingly, one KPC/Y mouse retained MRTX113 sensitivity even after tumor regrowth; however, further studies with larger cohorts are needed to determine whether this is a widespread property.

Figure 1.

MRTX113 treatment leads to regression of KRASG12D-driven PDAC and alters the tumor microenvironment (TME). Treatment of KRASG12D PDAC cell lines with the selective inhibitor MRTX113 results in the inhibition of RAS signaling, as evidenced by a reduction in the levels of phosphorylated ERK1/2. The functional effects of this inhibition translate in vivo, as the administration of MRTX113 in KRASG12D-driven tumors not only increases apoptosis and blocks tumor cell proliferation, but also induces multiple changes in the TME. These changes include an increase in the M1/M2 macrophage polarization ratio, an increase in myofibroblastic cancer-associated fibroblasts (myCAF), an increase in expression of MHC class I on tumor cells, and increased CD4+ and CD8+ T-cell infiltration along with increases in the cytotoxicity marker GZMB. Both the intracellular effects of inhibiting KRASG12D and the altered TME likely contribute to the observed increase in cancer cell death and regression rates seen in MRTX113-treated tumors.

Figure 1.

MRTX113 treatment leads to regression of KRASG12D-driven PDAC and alters the tumor microenvironment (TME). Treatment of KRASG12D PDAC cell lines with the selective inhibitor MRTX113 results in the inhibition of RAS signaling, as evidenced by a reduction in the levels of phosphorylated ERK1/2. The functional effects of this inhibition translate in vivo, as the administration of MRTX113 in KRASG12D-driven tumors not only increases apoptosis and blocks tumor cell proliferation, but also induces multiple changes in the TME. These changes include an increase in the M1/M2 macrophage polarization ratio, an increase in myofibroblastic cancer-associated fibroblasts (myCAF), an increase in expression of MHC class I on tumor cells, and increased CD4+ and CD8+ T-cell infiltration along with increases in the cytotoxicity marker GZMB. Both the intracellular effects of inhibiting KRASG12D and the altered TME likely contribute to the observed increase in cancer cell death and regression rates seen in MRTX113-treated tumors.

Close modal

The model-dependent differences in tumor regression, although not surprising given the well-established context-dependent differences in oncogenic RAS signaling and the tumor microenvironment (TME) of PDAC, provide important insight into the mechanism(s) of action of MRTX113. A major obstacle in treating KRAS-mutant PDAC stems from its dense, hypovascular, and immune-poor TME. Mutant KRAS itself has been implicated in maintaining this immunosuppressive environment by inhibiting antigen presentation, blocking interferon signaling, and increasing the levels of myeloid chemoattractants (9). Consistent with these findings, KRASG12D inhibition by MRTX113 was accompanied by drastic changes in the PDAC TME (Fig. 1). A short treatment with MRTX113 (5 doses, over 60 hours) resulted in a reduction in total myeloid cells, granulocytic myeloid-derived suppressor cells, and dendritic cells, as well as an increase in macrophages and a shift toward an M1 state in the subcutaneous PDAC models. Supporting these observations, MRTX113 induced changes in cytokine profiles that would promote antitumor macrophage infiltration and inhibit myeloid-like macrophage infiltration, such as increases in CCL2 and decreases in GM-CSF. MRTX113 treatment also resulted in a significant increase in tumor-infiltrating T cells (CD3+) and CD4+ and CD8+ T-cell subsets, with CD8+ T cells expressing higher levels of proliferation and cytotoxicity markers (Fig. 1). In contrast to macrophages, however, the differences in T cells were not sustained with longer treatment. These data suggest that MRTX113 treatment could help transition immune-cold tumors into tumors with some degree of immune infiltration. However, they also indicate that the effect of KRASG12D inhibition on the PDAC immune microenvironment can be dynamic and as such must be carefully considered in the design of approaches that combine immunotherapy with KRASG12D inhibition. Interestingly, the degree of regression appears to vary depending on the immune infiltrate status of PDAC. When treated with MRTX113, 100% of immune-hot (high T-cell infiltrates) subcutaneous tumors regressed, with half of these being CR. Meanwhile, only 50% of immune-cold tumors regressed, and no CR was observed. The combination of MRTX113 with T-cell depletion in the immune-hot tumors had lower efficacy than MRTX113 alone, as only 86% of the tumors regressed and no CR was observed, suggesting that T cells play a role in the antitumor effect of MRTX113 in this model. However, the depletion of T cells in the autochthonous KPC model had no impact on regression, as these models experienced a 100% regression rate with or without T-cell depletion. The role of T cells in mediating the activity of MRTX113 may therefore be context (site) specific. Nonetheless, when T cells were continually depleted after MRTX113 therapy was stopped in immune-hot subcutaneous tumors, 100% of tumors showed substantial regrowth, whereas 50% of tumors with functioning T cells remained in remission. Depletion of T cells also exacerbated regrowth in KPC/Y tumors after MRTX113 therapy. Thus, T cells may play an important role in the durability of response to MRTX113. Finally, surviving tumor cells after MRTX113 treatment showed an increase in MHC class I. This reexpression was similarly seen with KRASG12C inhibition, leading to newfound sensitivity to immunotherapeutic intervention (10). Alterations in the additional components of the PDAC TME may also contribute to the antitumor effect of MRTX113 and changes in the immune landscape. As such, MRTX113-treated tumors showed elevated collagen deposition paralleled by an increase in myofibroblastic cancer-associated fibroblasts, which have been shown to be tumor-suppressive. In the subcutaneous models, tumor vascularization also increased after MRTX113 treatment, but this was not observed in the orthotopic setting.

Overall, the identification of MRTX113 generates immense potential for the treatment of patients with KRASG12D-driven PDAC. Not only is it the first of its kind, but the experimental evidence provided demonstrates that MRTX113 is a potent inhibitor of KRASG12D with unprecedented antitumor activity in preclinical models of PDAC. Although key questions regarding the hierarchy and interplay of KRAS downstream signaling pathways involved in the antitumor effect, durability of response, and resistance mechanisms following MRTX113 treatment must be considered for optimal use in patients, these findings are a great leap forward in the treatment of KRASG12D-driven PDAC.

No disclosures were reported.

This study was supported by an NIH/NCI T32 grant in Cancer Biology (T32CA236736) to A. Redding and an NIH/NCI R37CA230645 grant to E. Grabocka.

1.
Waters
AM
,
Der
CJ
.
KRAS: the critical driver and therapeutic target for pancreatic cancer
.
Cold Spring Harb Perspect Med
2018
;
8
:
a031435
.
2.
Ostrem
JM
,
Peters
U
,
Sos
ML
,
Wells
JA
,
Shokat
KM
.
K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions
.
Nature
2013
;
503
:
548
51
.
3.
Janne
PA
,
Riely
GJ
,
Gadgeel
SM
,
Heist
RS
,
Ou
SI
,
Pacheco
JM
, et al
.
Adagrasib in non-small-cell lung cancer harboring a KRAS(G12C) mutation
.
N Engl J Med
2022
;
387
:
120
31
.
4.
Hong
DS
,
Fakih
MG
,
Strickler
JH
,
Desai
J
,
Durm
GA
,
Shapiro
GI
, et al
.
KRAS(G12C) inhibition with sotorasib in advanced solid tumors
.
N Engl J Med
2020
;
383
:
1207
17
.
5.
Wang
X
,
Allen
S
,
Blake
JF
,
Bowcut
V
,
Briere
DM
,
Calinisan
A
, et al
.
Identification of MRTX1133, a noncovalent, potent, and selective KRAS(G12D) inhibitor
.
J Med Chem
2022
;
65
:
3123
33
.
6.
Kemp
SB
,
Cheng
N
,
Markosyan
N
,
Sor
R
,
Kim
IK
,
Hallin
J
, et al
.
Efficacy of a small-molecule inhibitor of KrasG12D in immunocompetent models of pancreatic cancer
.
Cancer Discov
2023
;
13
:298–311.
7.
Lee
JW
,
Komar
CA
,
Bengsch
F
,
Graham
K
,
Beatty
GL
.
Genetically engineered mouse models of pancreatic cancer: the KPC model (LSL-Kras(G12D/+);LSL-Trp53(R172H/+);Pdx-1-Cre), its variants, and their application in immuno-oncology drug discovery
.
Curr Protoc Pharmacol
2016
;
73
:
14.39.1
20
.
8.
Hallin
J
,
Bowcut
V
,
Calinisan
A
,
Briere
DM
,
Hargis
L
,
Engstrom
LD
, et al
.
Anti-tumor efficacy of a potent and selective non-covalent KRASG12D inhibitor
.
Nat Med
2022
;
28
:
2171
82
.
9.
Mugarza
E
,
van Maldegem
F
,
Boumelha
J
,
Moore
C
,
Rana
S
,
Sopena
ML
, et al
.
Therapeutic KRAS(G12C) inhibition drives effective interferon-mediated antitumor immunity in immunogenic lung cancers
.
Sci Adv
2022
;
8
:
eabm8780
.
10.
Zhang
Z
,
Rohweder
PJ
,
Ongpipattanakul
C
,
Basu
K
,
Bohn
MF
,
Dugan
EJ
, et al
.
A covalent inhibitor of K-Ras(G12C) induces MHC class I presentation of haptenated peptide neoepitopes targetable by immunotherapy
.
Cancer Cell
2022
;
40
:
1060
9
.