ArticlePDF AvailableLiterature Review

ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework

Authors:

Abstract and Figures

At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Content may be subject to copyright.
npj | precision oncology Review article
Published in partnership with The Hormel Institute, University of Minnesota
https://doi.org/10.1038/s41698-024-00554-5
ERK pathway agonism for cancer therapy:
evidence, insights, and a target discovery
framework
Check for updates
Oleg Timofeev1,PhilippeGiron
2, Steffen Lawo 3, Martin Pichler4& Maxim Noeparast 4
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors
targeting various effectors within the ERKp have been developed and explored for over two decades.
Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a
greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this
vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on
the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds.
We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for
therapeutical approaches and contextualize these insights within established pharmacological
cancer-targeting models. Moreover, we compile the intriguing preclinical ndings of ERK pathway
agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery
regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to
develop novel targeted therapies for precision oncology.
Background
Cancer targeting: from chemotherapy to precision oncology
In 1904, after a series of studies on different model organisms, Paul
Erlich, a highly meritorious German medical scientist, coined che-
motherapy to address his approach when chemicals were used to target
infectious diseases1,2(see Fig. 1). Ehrlich humbly stated: Indeed, from the
very origin of the art of healing, chemotherapy has existed since almost all
the medicaments we employ are chemicals1. However, Ehrlichs concept
of chemotherapy unprecedently linked the chemical structure and
chemoreceptors on the target cells to make sense of the pharmacological
activity3,4. He address ed chemicals that selectively exert their detrimental
effect on the target cells (e.g., parasites) and not on the treated organism
cells as Zauberkugeln or magic bullets1,2. Erlich had tested compounds,
such as alkylating agents, to target cancer, which, in his hands, showed
limited effects, far from being considered Zauberkugeln. Still, Ehrlich
remained optimistic that effective chemotherapeutics could be explored
in the future1. He explained this lack of effect by the high similarity
between cancer and non-cancer cells1.
During World War II, in a mouse xenograft lymphoma model, two
American pharmacologists, Alfred Gilman, and Louis Goodman, showed
that nitrogen mustard, a warfare gas, had (chemo)therapeutic effects5,6.
Their work immediately justied the treatment of a nonHodgkinslym-
phoma patient and other cancers with nitrogen mustard5,7.Despitefurther
knowledge about its limited efcacy and proneness to resistance, nitrogen
mustards ephemerous success laid the foundation for establishing cancer
chemotherapy as a valid eld. Since then, several cancer chemotherapeutics
such as Alkylating, Antimicrotubular agents, and Antimetabolites have been
developed8. Cancer chemotherapy is a term whose denition was progres-
sively elucidated long after it was coined, considering the expanding
knowledge about chemotherapeuticsmechanism of action and limitations.
Compared to more selective therapeutics, Chemotherapeutics are con-
sidered one-size-ts-all treatments targeting different cancer types carrying
distinct driver oncogenes8. Despite their narrow therapeutic window and the
possibility of targeting non-cancerous dividing cells, chemotherapeutics
have saved lives in some cancer populations, such as pediatric hematologic
cancers, or among adult malignancies, such as testicular cancer9.Until
1Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043 Marburg, Germany. 2Vrije Universiteit Brussel
(VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics,
Laarbeeklaan 101, 1090 Brussels, Belgium. 3CRISPR Screening Core Facility, Max Planck Institute for Biology of
Ageing, 50931 Cologne, Germany. 4Translational Oncology, II. Med Clinics Hematology and Oncology, 86156
Augsburg, Germany. e-mail: maxim.noeparast@gmail.com
npj Precision Oncology | (2024) 8:70 1
1234567890():,;
1234567890():,;
today, chemotherapy, surgery, and radiotherapy are on the list of available
neoadjuvant, adjuvant, or combined cancer treatments8,10.
Followed by the burst of knowledge about cancer-related gene muta-
tions in the 1990s, targeting oncogenes laid the foundation for targeted
therapy in cancer11. Indeed, since the New Era of Personalized Medicine in
199911, cancer treatment has been revolutionized. We have seen the main-
stream shift from onesizetsallapproaches, such as classical che-
motherapy, to individualized treatments according to patientstumor
genetic proles12,13. A milestone in personalized cancer therapy was the
success of Imatinib, a small molecule inhibitor of the BCRABL protein
tyrosine kinase. This inhibitor was initially conceptualized and proled by
Nicholas Lydon and colleagues14. Imatinib demonstrated remarkable clin-
ical advantages during phase II trials, particularly among patients with
chronic myeloid leukemia (CML)15,16.
By 2002, to explain mechanisms behind the sensitivity of some cancers
to mutation-specic targeting, Bernard Weinstein coined the term oncogene
addiction to describe a phenomenon that, despite extensive genetic altera-
tions, cancer cells can depend on a single oncogene activity1723.Onecan
analogize eliminating that very oncogene activity in the addicted cell to
surpassing the cells system biology threshold, ultimately leading to a lethal
trade-off favoring proapoptotic vs. the prosurvival signals. Various ther-
apeutics have been developed against other oncogenes to which different
cancers are addicted. EGFR- or ALK-altered lung cancers and BRAF mutant
melanoma are some success stories of personalized cancer treatments2426.
All these therapeutics work by inhibiting a driver oncogene or its down-
stream effector, based on the rationale that withdrawal from oncogene
addiction leads to deleterious oncogenic shock27. Indeed, target inhibition is a
shared feature of all these treatments. Although the targets are often kinase,
non-kinase protooncogenes, such as KRASG12C, are also actionable28.Sooner
or later, however, all these treatments are doomed to the rise of resistance
mechanisms, leading to the relapse of the treated cancer25,2933.
A theoretical alternative of oncogene inhibition as a therapeutic con-
cept is the re-introduction or restoration of a lost tumor suppressor (TS)
activity, as cancer cells might be sensitive to that lost function34,35. However,
this strategy has been proven challenging as often, if not always (e.g., some
p53 variants), TS genes are lost due to various genetic and epigenetic
alterations and, therefore, non-targetable in cancer cells. The p53 targeted
therapies, conceptualized according to the p53 status of the cancer cell, are
being explored36. These approaches can involve different strategies, from
stabilizing the wild-type p53 and unleashing its wide range of TS functions
in cells with existing wild-type copies to restoring the wild-type con-
formation among responsive mutant p53 variants36. In the bargain,
numerous efforts have been made to re-express tumor suppressors by viral
and non-viral gene therapy in tumor cells37.Viraltumorsuppressorgene
transfer has even been combined with the TS-loss-targeted-oncolytic
capacity of therapeutic viruses or synergistic effects of TS re-expression with
immunotherapeutics to double-punch the cancer cells3740.Despiteallthese
cumbersome efforts, none has yet borne fruit in the clinic among large
patient populations. Moreover, in the late stages of cancer, due to the highly
divergent genetic build-up of late-stage tumor cells, it is to be further
elucidated whether cells have already circumvented their native sensitivity
and response to the lost tumor suppressors.
By 1945, Theodosius Dobzhansky, a Russian-American geneticist, had
coined the term synthetic lethality, describing a scenario when two dis-
tinctive chromosomes in Drosophila pseudoobscura were tolerated if exist-
ing alone but became lethal when they co-existed synthetically, as being
imposed in the lab, in cross-over ies41. In 2009, Nobel Prize laureate
William Kaelin Jr. put synthetic lethality forward as a conceptual framework
for exploring novel anticancer treatments42. The cancer research eld fur-
ther translates this as the genetic build-up in certain cancers conferring
sensitivity to specic chemical or genetic perturbations, opening novel
avenues for indirectly targeting previously non-druggable targets43,44.Today,
synthetic lethality is described as the cellular lethality of at least two co-
occurring contextual (genetic) or chemical perturbations targeting at least
two separate genes. In contrast, these perturbations are tolerated at
variance45. As a result, within the scope of personalized medicine, treatments
have been developed by targeting specic non-oncogenes based on the
concept of synthetic lethality43,44. An example of such an approach in the
clinic is the adjuvant as well as palliative PARP inhibitor treatment of
BRCA1/2-mutant HER2-negative breast cancer46.Anexcitingfeatureof
synthetic lethality is that it can be applied to both an oncogene and a tumor
suppressor as long as they confer sensitivity to a synthetically lethal target44.
An interesting reconceptualization of synthetic lethality as a cancer-
targeting approach, namely the one-two punch47 model, is being pioneered
and explored by René Bernards and colleagues. In this model, the rst-line
treatment is designed to induce senescence-associated vulnerabilities in
cancer cells that are to be targeted with the second-line senolytic
compounds47.
Notably, in recent years, targeting cancer-related immune cells or their
interaction with cancer cells has opened a new front in the battle against
cancer4850. For instance, in BRAF mutant melanoma, in which targeted
inhibitors are known to be at their highest performance, immune check-
point inhibitors showed a 20% benet over targeted compounds in terms of
overall survival among therapy-naïve and metastatic patients51.Assuch,
oncogene-targeted therapy might seem on the verge oflosing momentum in
personalized medicine. However, despite the striking responses to different
cancer immunotherapeutics in a group of patients, many patients remain
non-responsive or fast-resistant to such therapies4850,52.
While it is essential to work towards enhancing existing therapeutic
strategies, it is also highly justied to put forward and explore innovative and
unconsumed concepts.
By 2015, two signicant studies showed that mutual exclusivity among
BRAF/KRAS and EGFR/KRAS oncogenes can result from synthetic leth-
ality or senescence43,53.
The British fairy tale Three Bears(Eleanor Mure, 1831) revolves
around a greedy individual who, in the absence of the three bachelor bears,
each of varying sizes, enters their cottage54. She samples their trio of distinct
milk portions, chairs, and beds. Repeatedly,amidstthesetriads,shedis-
covers contentment solely when the element is precisely balanced not too
much, not too little, but just right54. In later biological and medical contexts,
Fig. 1 | Timeline of cancer-targeting discoveries and concepts relevant tothis writi ng. The focus of the timeline is mainly on therapeutics targeting the cancer cells. Figure
generated in Biorender.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 2
the widely spread but modied version of the original story Goldilocks and
the Three Bears,in which a young girl replaces the elderly woman, has
inspired the Goldilocks principle. This principle signies that, for a biological
system to function optimally, its components should possess the correct
levels of abundance and activity and be temporally right avoiding both
excess and deciency55.
Amit Dipak Amin and Jonathan Schatz, inspired by the Goldilocks
principle in Biology, introduced the term and the concept of Oncogene
Overdose.This concept compares cancer cells to drug addicts, as both can
die due to an overwhelming excess of what they are addicted to55.
In 2016, Nobel laureate Harold Varmus introduced the concept of
harnessing mechanisms elicited by the co-induction of mutually exclusive
genes as a therapeutic model56.Furthermore,Xuetal.fromDengs
laboratory have explored a striking KRAS agonist for targeting KRAS
mutant cancers57. More recently, Sugiura et al. presented the therapeutic
concept of ERKp activators5860. Concurrently, René Bernards and collea-
gues described Paradoxical Intervention, signifying the simultaneous acti-
vation of mitogenic signals and the employment of stress-inducing
compounds in cells with pre-existing ERK pathway-activating
alterations61,62. Remarkably, they have demonstrated that such treatment
enforces unprecedented tumor-suppressive resistance mechanisms62.
Dostoevsky once remarked, We all come out from GogolsOvercoat,
paying homage to Nikolai Gogol as the iconic predecessor of Russian lit-
erature preceding his own generation. In doing so, he employed wordplay by
referring to one of Gogols renowned short stories, namely, The Overcoat.’“
Based on chronological order and logical reasoning, all the above concepts
revolve around over-activating the already activated pathway as a ther-
apeutic approach, akin to stepping out from oncogene overdose overcoat.
In this writing, we review the evidence and insights that reinforced ERK
pathway effectors activity in cells with aberrantly increased ERK activity is
damaging to these cells. The pathway is also known as mitogen-activated
protein kinase (MAPK) or Ras-Raf-MEK-ERK pathway. In this writing, for
simplicity, we will mention the pathway by its major effector ERK without
distinguishing between the two human ERK genes, ERK1 and ERK2. Fur-
thermore, we will present a conceptual framework for therapeutic targeting
of such vulnerabilities.
The double-life of ERKp in human cancer and the curious case of
the ERK proteins
The ERK pathway is one of the extensively studied signal transduction
cascades. Essential effectors of this signaling cascade in humans fall into
three types of eukaryotic protein kinases: (1) HER family Receptor Tyrosine
Kinases (RTKs, EGFR, HER2-4), RAF family serine/threonine kinases (A/
B/CRAF), and (3) dual specicity protein kinase families MEK (MEK1/2)
and ERK (ERK1/2)6365. In humans, the RAS family, consisting of three
essential members, H/N/KRAS, serve as the principal non-kinase effectors
within the ERK pathway66.
Feedback loops regulate ERK signaling during development and in
normal physiologic conditions for cell proliferation, survival, and
homeostasis6366.
Due to genetic alterations of the ERK pathway effectors and/or its
regulatory components, ERK signaling may become pathologically
deregulated. Aberrant down-regulation of the ERK pathway can be asso-
ciated with some neurodegenerative or autoimmune disorders, while its
abnormal upregulation is associated with human cancers, rasopathies, and
Erdheim-Chester disease6669.
Under normal cellular conditions, upon ligands binding, HER recep-
tors can undergo conformational changes, transitioning to an active state
and forming homo- or hetero-dimers70. The induced conformational
alterations in the tyrosine kinase domain of dimerization partners facilitate
ATP binding in their ATP binding cleft and, as suggested, in the case of
EGFR, might lead to the relief of cis-autoinhibition and trans-
autophosphorylation of the tyrosine kinase domains70,71.
Under physiological circumstances, the mature RAS protein, when
residing in the inner surface of the plasma membrane, undergoes
conformational changes mediated by the upstream HER signaling or reg-
ulatory feedback signals66,72. RAS alternates between a guanosine tripho-
sphate (GTP)-bound state and a guanosine diphosphate (GDP)-bound
state28,66,73. The oscillation of RAS into its active conformation involves GTP
loading28,66,73. This process can be initiated by recruiting GRB2 proteins to
the plasma membrane, where the GRB2 is activated via its SH2 domain by
an active RTK66. Subsequently, SOS is recruited, forming a complex invol-
ving at least GRB2, SOS, and RAS66. This complex facilitates the displace-
ment of GDP and promotes the loading of GTP onto the RAS molecule66.
RAS can hydrolyze its GTP as a GTPase protein, converting it to GDP and
becoming inactive28,66,73. This process is facilitated by GTPase-activating
proteins (GAPs)28,66,73. Feedback loops play a regulatory role in the GTPase
activity of RAS66. The GTP-bound RAS triggers the activation of down-
stream RAF by engaging in a physical (allosteric) interaction with RAF or
mediating the release of its autoinhibition74. RAFs, in turn, phosphorylate
the MEK1/2 through phosphotransferase activity, and MEKs, in turn,
phosphorylate the ERKs, which, due to the Erksvast array of network,
majorcellulareventssuchasproliferationandsurvivalcantakeplace
6366.At
least 659 of ERKsdirect substrates are discovered75.
Approximately 40% of human cancers are linked to the abberant
upregulation of ERKp76. Notably, cancers that harbor genetic alterations in
an ERKp effector can be associated with worse clinical outcomes than
cancers where such alterations are absent, emphasizing the signicance of
these alterations on cancer prognosis77. For over two decades, the effective
inhibition of the ERKp has remained a key focus in cancer research and
targeted therapy efforts12,24,26,28,77.
However, in an ironic twist, a wealth of evidence underscores the re ality
that human cells, including cancer cells, cannot tolerate excessive levels of
ERKp activity. While upstream effectors of the ERKp, such as RTKs, RAS,
and RAFs, are subject to activating and oncogenic mutations in humans, the
mutations in downstream effectors, such as MEK and even to a greater
extent ERK, are infrequent26,73,77,78. Before the ERK small molecule inhibi-
torsdevelopment and clinical employment, ERK mutations were hardly
reported in human cancer79. These mutations can be associated with sec-
ondary resistance mechanisms that lead to loss-of the protein afnity to
ERKinhibitorsincellstreatedwiththesecompounds
7981. As illustrated in
Fig. 2, among the conventional ERK signaling pathway effectors, the ERK
mutations still rank at the bottom of the list regarding the frequency of
mutations in human cancer.
The ERKs need two critical phosphorylation events before full con-
formational activation, none triggered by either cis- or trans-
autophosphorylation mechanisms82.AsopposedtoRTKs,RAFs,oreven
MEKs, ERKs protein conformations have evolved in such a way to be less
poised for autoactivation and instead rely on direct phosphotransferase
activity of MEKs for full activation82,83.
Several activating variants that cause increased kinase-dependent or
-independent activity of the ERK orthologues, such as Sevenmaker variants,
are studied in Yeast and Drosophila models8385. However, how such
mutations can induce or contribute to human carcinogenesis remains
enigmatic. Several investigations have concentrated on characterizing a few
purportedly activating ERK variants by expressing them in mammalian
cells82,86,87. These studies have looked into activating variants of ERK that
render the ERK protein an autoactivation capacity82,86,87. In particular, the
activating variant ERK1R84H, reported twice in human cancer, was found to
transform NIH3T3 cells87. While introducing these presumably activating
ERK variants into non-cancerous mammalian cells was occasionally fea-
sible, accomplishing the same in human cancer cell lines appeared to pose
challenges. Markedly, Goetz et al. had shown in the past that overexpression
of wild-type ERK1/2 and not the kinase-dead or low-kinase ERK1/2 variants
in BRAFV600E mutant melanoma cell line A-375 leads to growth inhibitory
effects88. Interestingly, the ERK1/2 variants with increased kinase activity,
which were found to confer resistance against RAF inhibitors (RAFi) and
MEK inhibitors (MEKi) during a random mutation screen, even exerted
more potent growth inhibition when overexpressed in A-375 cells. The most
active ERK1/2 variants exhibited growth-suppressive effects in two other
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 3
BRAF mutant melanoma cell lines beyond A-375 cells(SKMEL-19, and
WM266.4)88. It was also shown that inducible overexpression of ERK2 in
A-375 cells was selectively detrimental to these cells vs. cells with wild-type
BRAF and caused anti-tumor effects in vitro and in vivo89. Such detrimental
effects could only be rescued uponERK2orBRAFknock-down
89.The
ERK2 overexpression in these cells was associated with the induction of ER
stress and DNA damage in addition to proapoptotic signals89.Thefactthat
some ERK-associated cancers, such as BRAF mutant melanoma, are sen-
sitive to ERK activation is well-established.
Interestingly, the negative effect of putative Gain-of-Function
(GOF) mutations on the proliferation of these cells has been utilized as a
model in an ERK saturation mutagenesis study90. Such a comprehensive
approach by Brenan et al. has shed light on the relevance of ERK
mutations in human cancer. It is worth noting that saturation muta-
genesis of ERK was unsuccessful in identifying the direct GOF impact of
activating variants, unless upon MEKi and BRAFi treatment. Indeed,
expression of GOF sevenmaker ERK mutations ERK2D321N and
ERK2E322K or other supposedly GOF variants in the A-375 cell line had a
robust anti-proliferative effect in these cells90. Interestingly, the seven-
maker variant and thos e that phenocopy its effect could only be tole rated
by the BRAFV600E cells under MEK or RAF inhibition90. These variants
are proposed to enhance ERK activity by disrupting ERKs interaction
with inhibitory DUSP phosphatases90.
The rarity of ERK mutations in human cancer raises an intriguing
question, prompting consideration of at least two alternative scenarios to
explain this phenomenon.
One possibility is that the ERK protein, by default, exhibits low basal
activity and has a weak impact on its network. Consequently, even an
activating mutation might not be sufcient to manifest an effective GOF
phenotype leading to cell transformation. An illustrative example of such a
scenario is the case of CRAF mutations in human cancer, which are rarer
than mutations of another RAF isoform, BRAF91,92.Pastexplanations
attribute this rarity to the low basal activity of CRAF compared to BRAF93.
Conversely, the effectors, such as EGFR,whicharemoreupstreamandhave
a wider array of targets belonging to distinctive pathways, exhibit a higher
frequency of mutations in human cancer. From a vertical signaling cascade
standpoint, the three commonly mutated effectors of the ERKp in human
cancer are arranged as EGFR, KRAS, and BRAF. EGFR and BRAF muta-
tions exhibit a comparable frequency in human cancer (see Fig. 2). Notably,
KRAS mutations occur at a frequency equivalent to the combined occur-
rences of EGFR and BRAF oncogenes (Fig. 2). The higher mutational fre-
quency of KRAS over BRAF can be explained by its vertical rank along the
signaling cascade, a rationale not applicable to the other two effectors82.
Another explanation could be that while ERK mutations capable of
generating an effective GOF phenotype may exist synthetically, they are
negatively selected in the real world due to being poorly tolerated by human
Fig. 2 | ERK1/2 mutations are relatively rare. In July 2023, the query of
69223 samples belonging to 65853 patients in 213 curated and non-redundant
studies in cBiportal for only mutations among conventional effectors of the ERKp
yielded the approximate frequencies as displayed. Note that this oversimplied
schematic is not meant to communicate complex signaling and structures of the
effectors. Unlike typical oncogenes, ERK1/2 mutations are distributed along the
ERK proteins conserved protein kinase and non-conserved regions, with no hotspot
mutational site. Figure generated in Biorender.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 4
cells. In contrast to, for instance, KRAS, which oscillates between active and
inactive states under normal physiological conditions and may become
constitutively active in human cancer73, human cells cannot tolerate ERKs in
a constitutively active state. It is crucial to distinguish between two types of
ERK mutations. The rst type comprises mutations that occur in the real
world. As the above evidence suggests, these mutations exhibit low trans-
forming capacity. Then comes the synthetic ERK mutations, which do not
occurintherealworld.Indeed,asmentionedintheaboveparagraphs,such
synthetic variants have been studied in the past, and it has been shown that
they are not easily tolerated by human cells, particularly cancer
cells79,81,82,8486,8890. More interestingly, in addition to some ERK mutants,
even wild-type ERK overexpression is not tolerated in such human cellular
models88,90. Indeed, one of the vital pieces of evidence is ERK saturation
mutagenesis by Brenan et al., as they observed in human melanoma cell line
model with BRAFV600E, expression of wild-type ERK and activating ERK
variants could be tolerated only in the presence of ERK pathway inhibitors90.
This evidence, at least, can rule out the possibility that ERK, by nature,
cannot render gain-of-function. The scenario that ERK-activating muta-
tions are not easily tolerated by human cells aligns with one aspect of ERK
protein activity autoregulation, namely its reduced potential for auto-
activation, in contrast to other ERK pathway effectors like EGFR82,83.The
ERK protein is shown to tolerate synthetic mutations that can render it
increased autophosphorylation and kinase activity to levels comparable to
ERK582.
The RTK/RAS/RAF pathway and its inhibitors exhibit a Janus-faced
immunomodulatory effect in cancer (reviewed here94). In brief, inhibition of
the ERKp can enhance the activity of associated T-helper cells and cytotoxic
T-cells, thereby potentially enhancing the immune response against cancer
cells94. Additionally, it can enhance dendritic cell activity, which plays a
crucial role in antigen presentation and immune activation. Conversely, this
inhibition may hamper tumor inltration of immunosuppressive regulatory
T-cells, monocytes, and macrophages, potentially limiting their immuno-
suppressive functions94.Ontheotherhand,long-termERKpinhibition
might eventually have an immunosuppressive effect95. Of note, oncogenic
RAS can contribute to immune evasion by stabilizing the PD-L1 mRNA and
subsequently favoring the PD-L1 expression on tumor cells96.
ERK pathway hyperactivation can be lethal to cells in
various ways
The ERKp activity can lead to cell toxicity and death (see Fig. 3). Numerous
studies have unraveled such a role in different model organisms60,97,98.The
ERK-induced cell apoptosis can involve extrinsic or intrinsic apoptotic
pathways60,97,99. ERK activity affects cell death by inuencing multiple cel-
lular processes, including mitochondrial dysfunction100102,DNADamage
Response103, Endoplasmic Reticulum stress104,Autophagymodulation
60,105,
Metabolic imbalance106, and accumulation of Reactive Oxygen
Species(ROS)97,103. ERKp activity can trigger senescence in vitro and
in vivo97. An interesting example of such a phenomenon is the existence of
benign human naevi with BRAFV600E mutation107,108. In cases where this
mutation occurs in isolation and without the concurrent loss-of the p53
tumor suppressor, it has been proposed to result in irreversible cellular
senescence rather than malignancy108. More recently, an alternative
mechanism involving non-coding RNAs has come to light, linking
BRAFV600E-carrying benign naevi to occurrences of mitotic failure and
reversible proliferation arrest107.
ERKp activity is crucial in determining divergent cellular outcomes,
including cell proliferation, growth arrest, or cell death. Notably, Hong et al.
from Parks lab have elucidated an exclusive threshold for ERK activity109.
Previously, it was known that excessive activity of ERKs, like other effectors
such as MEKs, CRAF, and BRAF, can cause growth arrest110113. However,
Hong et al. show that very high activity levels of ERKp, exceeding those that
cause growth arrest, can lead to apoptosis109. These ultra-high ERKp levels in
HEK293 and U251 cells could only be achieved by combining ERKs
overexpression and the presence of tamoxifen-inducible active CR3 catalytic
domain of CRAF109. As proposed by Hong et al., this cell death-inducing
characteristic is unique to the ERKs compared to the upstream kinase
effectors of the ERKp109.
Overall, the evidence is ample that excessive ERK activity is toxic to
cancer cells with different tissue backgrounds. First, we highlight a few
critical early studies among such evidence. Early evidence of ERKs
proapoptotic activity was observed in the human breast cancer cell line
(MCF-7), as RAF depletion could desensitize these cells to Paclitaxel114.
During the characterizationof Phenethyl Isothiocyanate anti-proliferative
effects against p53-decient prostate cancer cell line PC-3, ERKp pro-
longed activation was responsible for the compounds growth inhibitory
effects115. Moreover, as shown in mouse embryonic broblasts and MCF-
7, the DNA Damage (DD) caused by different stimuli, including Etopo-
side, could trigger ERKp activationindependent of p53 but rest on Ataxia-
Telangiectasia Mutant (ATM)116. The ERK activity was essential for DD-
triggered growth arrest and apoptosis116. Later, it was shown that growth
inhibitory and proapoptotic effects of Asiatic acid against breast cancer
cell lines (MCF-7 and MDA-MB-231) were associated with the induction
of ERKp117. In another study investigating Lauryl-gallates growth-
suppressive and proapoptotic effect on three breast cancer cell lines MCF-
7, MCF-7 ADR, and MDA-MB-231, the observed phenotype was
accompanied by ERKp activity and p21-induced Cell Cycle (CC) arrest118.
As one of the investigated cell lines was p53 mutant (MDA-MB-231) and
the other possessed a multidrug-resistant phenotype (MCF-7 ADR),
authors conclude that none of these conditions affects the sensitivity of
cells to the tested compound118. Interestingly, MEK inhibition and the
resulting ERKp suppression were associated with rescuing the drug
effect118. In another study, sustained ERKp activity due to exogenous
expression of active MEK1/2 led to G1 CC arrest, marked by p21WAF
expression119. ProlongedERK activity was associated with the activation of
cellular protein biosynthesis regulator p70S6K, accompanied by an
increased translation of p21 protein120. Nonetheless, none of these studies
has fully elucidated the mechanisms elicited by ERK activation to lead to
the observed phenotypes. Several other studies demonstrate that rein-
forced ERK activity in cancer cells potentiates the cytotoxic effects of
various chemotherapeutical agents (reviewed here60,97). In many of these
studies, the MEK inhibitor compounds, if among other means, were
employed to investigate the rescue effects60,97. Moreover, as recently
highlighted by Sugiura et al.60 and taking into account subsequent reve-
lations regarding MEK inhibitors off-targets and mechanisms of action,
these factors could potentially undermine the precision of the above-
mentioned ndings.
A revisit of the precedent evidence reveals an intriguing aspect of
ERKp-induced cell death and senescence, as it can happe nin both p53 wild-
type and p53-altered backgrounds114120. Numerous studies have indicated
crosstalk and interactions between the p53 and ERKp, suggesting that they
can inuence each others functions and responses under specic cellular
conditions. As a major tumor suppressor known as the guardian of the
genome121,p53 governs safeguard mechanisms that prevent the accumu-
lation of DNA damage and the proliferation of damaged cells122.Thep53
protein primarily functions as a transcription factor, regulating a plethora of
genes that control cell proliferation, apoptosis, DNA repair, and other cel-
lular processes123. Additionally, p53 possesses multiple transcription-
independent activities in cell death124126, metabolism127,128,autophagy
129,
DNA replication130, and repair131, all of which contribute to tumor sup-
pression and the maintenance of genomic integrity. The aberrant activation
of the ERKp leads to the induction of the p53 response, which can trigger
senescence108,132136 or cell death109,137. The classical mechanism of p53 acti-
vation by oncogenes is mediated by the p14ARF protein, which sequesters
the p53 inhibitory ubiquitin ligase Mdm2 (HDM2 in humans), allowing for
the accumulation of the p53 protein138. Additionally, studies have shown
that ERK can phosphorylate p53 on Ser15 under stress conditions, inhi-
biting its interaction with Mdm2 and promoting the stabilization of the p53
protein139141. The phosphorylation of p53 by ERK2 on Thr55 was reported
to induce p53 stabilization and activation in doxorubicin-treated MCF-7
cells142. However, it remains unclear to what extent the direct
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 5
phosphorylation of p53 by ERK contributes to the induction of senescence
and apoptosis in tumor cells without additional stress stimuli.
In the process of malignant transformation, cells with hyperactive ERK
signaling experience strong negative selective pressure from p53132,143149
which they can counteract by blunting p53 activity, often through the
acquisition of TP53 mutations that entirely or partially inactivate the tumor
suppressor150. Large-scale genomic studies have demonstrated that TP53
mutations are found in all types of cancers151. The frequency of TP53 genetic
alterations, estimated at an average of 50%, varies signicantly depending on
the tumor entity, ranging from 5% in neuroblastoma to over 90% in ovarian
and small-cell lung cancer151156.
Somatic mutations that inactivate TP53 are predominantly observed in
the later stages of tumorigenesis, indicating its role in advanced cancer
progression157. It is estimated that 80% of genetic alterations in the TP53
gene are missense mutations158,159 that lead to the expression of mutant p53
proteins. These mutants can possess oncogenic properties, driving cancer
progression and conferring therapy resistance160,161. However, the functional
impact of the majority of cancer-associated TP53 missense mutations
(approximately 70%) remains poorly characterized, and their role in
tumorigenesis and therapy response remains unclear162.
TP53 mutations oftenco-occur with oncogenicEGFR, BRAF,and RAS
mutations, but the extent of cooperation between TP53 and oncogenes may
vary even within the same tumor type. For instance, in lung adenocarci-
noma (LUAD), TP53 mutations are prevalent in EGFR mutant tumors but
are strongly underrepresented in KRAS-driven LUAD151,163, indicating
different paths of tumor evolution driven by different oncogenes. The
intricate interplay between various oncogenes within the ERKp and its
impact on the residual activity of wild-type or partial Loss-of-Function
(LOF) p53, retained in tumors, which can potentially lead to tumor-
suppressive effects, remains an area of ongoing investigation162,164.More-
over, different TP53 variants are suggested to cause distinctive LOF,
dominant negative, or even GOF phenotypes, which needs to be acknowl-
edged in further exploration of ERKp-induced lethality in different cellular
contexts with distinctive TP53 status165168.
ERK pathway in light of emerging knowledge about vulnerability
of cancers to replication stress targeting
In eukaryotes, transmitting genetic material to daughter cells is a crucial
event and is thus tightly regulated. Cyclin-dependent kinases (CDKs) are
pivotal in advancing the CC during interphase and M phases169171.TheCC
represents a series of sequential decisions and commitments169171.EachCC
stage is intricately governed by a complex interplay involving CDKs coupled
with E3 ubiquitin ligases like APC/C and their activating proteins169171.
Varied levels of CC-related proteins can establish decision windows that
impact entry, prevention of re-entry, or even the deceleration of CCs169171.
Although tightly controlled, the CC can face undesired events due to DD,
Replication Stress (RS), or spindle assembly malfunctions169171. Eukaryotic
cells rely on distinct checkpoints to tackle each scenario169171.RSemerges
when diverse factors slowdown or stall the replication fork. DD and RS
differ, but the latter can trigger DD as the collapse of the stalled fork can lead
to double-strand breaks169172. RS checkpoints function to avoid RS-induced
DNA damage response, while DD checkpoints are meant to prevent the
accumulation of DNA damage and thereby protect cells from subsequent
complications170. A cancers hallmark is uncontrolled proliferation, ham-
pering apoptosis and avoiding long-term CC exits34. Oncogenes like RAS
and MYC can negatively affect DNA replication licensing and ring,
Fig. 3 | Intertwining mechanisms by which excessive ERK activity leads to cell damage. Figure generated in Biorender.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 6
inciting RS169,170,172. Deregulation of DD and growth pathways due to cancer-
related mutations causes excessive S phase entry and subsequent RS. While a
temporary check-out from the Mitosis/entry is possible, leaving the CC is
not favored169172. Cancer cells hold a higher basal level of RS vs. normal
cells170. The severity of the damage and the context determines the cell fate,
which can be apoptosis, quiescence, or senescence. It deserves to be men-
tioned that DD checkpoint responses can lead to a temporary exit or, as
opposed to RS, an eternal exit of the CC169172. However, DD responses and
RS are so intertwined that, ultimately, RS can lead to such events169172.
Curiously, cancer cells highlight a distinct approach to DD and RS check-
points. DD checkpoints are often surrogated in cancer; cancer cells tolerate
defects in DD response and perhaps even benet from such a compromise
in favor of increased selection pressure, all while sidestepping unfavorable
exits from the cell cycle169172. Conversely, RS checkpoints are somehow
intact169172. Indeed, cancer cells are more sensitive to DD responses than RS
responses170. Cancer cells tolerate and even favor some Chromosomal
instability (CIN) levels170. In contrast, they cannot tolerate excessive CIN,
which can be caused by excessive RS and DD, leading to catastrophic mitotic
defects, loss-of-essential genes, and cell death170. Therefore, cancer relies on
RS checkpoints to avoid too much CIN. Consequently, targeting RS toler-
ance in cancer holds promise as a viable cancer therapy approach169173.Of
note, studies have demonstrated that elevated oncogenic RAS activity can
trigger RS by ubiquitously enhancing cellular transcription events174176.This
augmented transcription activity increases the likelihood of collisions
between replication and transcription processes174,176.InadditiontoRS
induction, oncogenic RAS (HRASG12V) can circumvent the p53 activity,
thereby sensitizing cancer cells to RS-inducing compounds176.
Withdrawing ERK pathway inhibitors from addicted cells: lethal
consequences of excessive pathway activity
There are several pieces of evidence that ERK-related cancer cells can tol-
erate upregulation of the ERKp only in the presence of ERKp inhibitors.
Resistance mechanisms against ERKp inhibitors predominantly involve
ERKp effectors and regulators30,31,81,177181. Different RAF and MEK inhibi-
tors can trigger clonal expansion of drug-tolerant cells, which maintain a
proliferative advantage, perhaps preferably in the presence of inhibitors by
virtue of enhanced ERKp activity182,183. This elevated activity could be lethal
upon inhibitor cessation, potentially resulting in ERK-related cellular
toxicities183. It is suggested ERKp inhibitors can create a window upon drug
removal, in which cells lose their tness advantage gained during drug
treatment and may even experience growth disadvantage due to excessive
ERKp activity and adaptive switching182,184. In an elegant study by Kong et al.
conducted in Peeper lab185, a CRISPR knock-out screen of melanoma cells
resistant and addicted to BRAFi revealed a phenotypic switch dependent on
ERK2 kinase and JUNB and FRA1 transcription factors accompanied by
suppression of microphthalmia-associated transcription factor (MITF)185.
The ERK2 dependency of the observed phenotype was supported by in vivo
and clinical ndings185. This dependency was also observed in lung cancer
cells resistant and addicted to EGFRi185. The addicted Melanoma cells
experienced grave cell death upon withdrawal from BRAFi, which could be
rescued by ERK2 targeting or restoring MITF activity185. Another intriguing
aspect of this study was that the authors opted to investigate the intermittent
drug treatment with the chemotherapeutic agent dacarbazine, and they
showed BRAFi-addicted melanoma cells were sensitized to this compound
accompanied with MITF inhibition185.
Aissa et al. elegantly showed at the single-cell level that drug-resistant
EGFR mutant lung cancer cell clusters exhibited markers indicative of
activated ERKp180. Recent work by Nuria Gutierrez-Prat et al.186 reported
similar ndings concerning ERK and MITF dependency on drug with-
drawal toxicity. Moreover, the authors nd that the knock-down of DUSP4,
an ERK phosphatase and negative regulator, was lethal by causing excessive
ERK activity186. This effect was observed not only in melanoma cells
addicted to inhibitors of the ERKp but also, intriguingly, in drug-naïve
cells186.Xueetal.inPiroLitos lab found the link between oncogenic BRAF
protein dosage in cells, the depth of ERKp inhibition and the related
resistance mechanisms. In their patient-derived xenograft lung cancer and
melanoma models, they discovered the more robust the ERK inhibition is,
the higher the oncogene dosage required for cells to retain proliferation
advantage in the presence of inhibitors183. They proposed a tness threshold
model, suggesting that cells treated with regimens with a higher threshold,
such as upon combination of RAFi, MEKi, and ERKi vs. ERKi mono-
therapy, might face a disadvantageous outcome due to sustained and
excessive ERKp activation when the drug treatment is stopped183.
Some preclinical and early clinical evidence suggested the benets of
intermittent treatment with RAFi and MEKi vs. sequential treatments, in
particular in melanoma182184,187197. This evidence laid the foundation for
clinical trials examining intermittent treatment regimens in individuals with
BRAF mutant melanoma198,199. Contrary to the expectations, these trials did
not show any overall survival benets from the intermittent therapies, and
even worse progression-free survival outcomes were reported upon inter-
mittent treatments198,199. Nevertheless, it is still uncertain whether those
intermittent therapies meant to produce that high tness threshold as
recommended by Xue et al. to cause selection disadvantages in tumor cells
during drug removal effectively.
Mutual exclusivity of highly activating variants of BRAF, KRAS
and EGFR oncogenes in cancer: induction of synthetic lethalityor
senescence
In 2006, Carlotta Petti et al.200 showed that synthetic expression of NRASQ61R
oncogene in a metastatic melanoma clone, which natively harbored the
mutually exclusive BRAFV600E oncogene, resulted in senescence. Later,
Cisowski et al. discovered that co-expression of BRAFV600E and KRASG12D
under their endogenous promotors provides a selective disadvantage
compared to single oncogene expression in mouse lung cells53. The decrease
in tumor burden in double oncogene-expressing tumors was associated with
hyperactivated ERK and AKT signaling and a decrease in proliferating
cells53. Further analysis demonstrated enhanced β-galactosidase expression
and increased p15, p16, and p19 levels upon oncogenes co-expression,
suggesting that double oncogene-expressing cells become senescent53.
Meanwhile, Unni et al. exogenously induced the expression of KRASG12V or
EGFRL858R in EGFREX19Del and KRASG12C LUAD cell lines, respectively43.
They observed decreased cell viability, indicating that mutant KRAS and
EGFR co-expression are not tolerated in cells43. Additionally, they generated
genetically engineered mice with co-induction of KRAS and EGFR mutants
in lung epithelium. The established lung tumors did not grow faster than
those harboring only one of the oncogenic mutations did43. Further analysis
indicated that only one of these oncogenes could be activated in the tumor
cells43. Furthermore, Unni et al. revealed that DUSP6 prevents ERK activity
from exceeding critical thresholds in EGFR and KRAS mutant cell lines201.
They found that targeting DUSP6 reduced cell viability due to unleashing
the excessive and toxic levels of RAS-mediated ERK activity in cancer cells
harboring mutations in EGFR and KRAS201. Markedly, Ambrogio et al.
showed that conditional induction of an EGFRL858R allele in KRASG12V
knock-in mouse LUAD models led to decreased tumor burden, increased
mice survival, and reversible cell toxicity in remaining tumor cells202.The
latter could further be recovered through ERKp activity reduction202.Of
note, all these consequences were associated with hyperactivation of ERKp
signaling43,201,202.
Harold Varmus, a Nobel Prize laureate, and his colleagues, who played
key roles in some of the aforementioned studies, articulated and cham-
pioned the intriguing concept that, unlike how previously considered, not
the redundancy of functions but synthetic lethality or senescence could lie
behind mutual exclusivity of certain oncogenes in cancer56.
We argue that the pathway redundancy and synthetic lethality,
senescence, or any other constraining phenomenon should not necessarily
be seen as conicting scenarios. One can consider that pathway redundancy
and synthetic lethality could both play roles in explaining mutual exclusivity.
When two proteins with overlapping functions in a pathway are excessively
active, the likelihood of both events being mutually exclusive within a cancer
cell is increased.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 7
On co-induction of activating EGFR and BRAF events in the same
cell, it has been shown that exogenous expression of wild-type EGFR in a
Melanoma cell line with native BRAFV600E leads to decreased proliferation
of these cells in a dose-dependent manner, in-vitro and in-vivo203.The
slowdown in proliferation was associated with cellular senescence as
suggested by hypophosphorylation of RB1 and induction of CDKN1A,
CDKN1B, and Beta-galactosidase203. Concerning mutant EGFR and
mutant BRAFs, two studies have been inspired so far by the emergence of
BRAFV600E in treatment-refractory EGFRL858R lung cancers as they become
resistant to EGFR-targeted therapies177,179. In one study, exogenous
expression of BRAFV600E in a polyclonal pool of EGFR mutant lung cancer
cells led to no differences in cellproliferation and cell death ratecompared
to empty vector control177. Of note, the BRAFV600E protein levels and
mRNA levels showed an indispensable increase only in the presence of an
ERKp inhibitor, suggesting that in the polyclonal pool, cells with
BRAFV600E induction are not clonally expanded unless the ERKp activity is
suppressed177. As such, further investigation is required to determine the
existence and the mechanism behind such clonal disfavor. Overall, the
ndings of thesetwo studies align with the results of two independent GOF
CRISPRa screens in Vemurafenib-treated A-375 cells (BRAFV600E),
showing that overexpression of EGFR, among others, is a resistance
mechanism to BRAFi204,205.
Three BRAF mutational classes26 include Class I variants, like
BRAFV600E, which often function independently of upstream effectors as
constitutively active monomers. Class II variants, exemplied by BRAFG469A,
activate the ERK pathway independently of RAS and CRAF as active
homodimers. Class III involves kinase-dead BRAF mutants activating the
ERK pathway through RAS-dependent allosteric transactivation of CRAF.
Cancer-relevant KRAS mutations are classied into three categories73
based on their impact on KRAS protein functions: Class I (Hydrolysis)
includes mutations leading to the loss-of the GTP-hydrolyzing feature of
KRAS, Class II (Exchange) involves mutations causing a gain in KRAS
Exchange function facilitated by Guanine Nucleotide Exchange Factors,and
Class III (Hybrid) encompasses mutations affecting both functions.
A recent classication of EGFR mutations24 considers both the struc-
tural effects of mutations on the EGFR protein, specically its drug-binding
pocket (DBP), and the implications of mutations on drug response.
Accordingly, one of the EGFR mutational classes is Classical-like (relevant
to this writing), where mutations like L858R have minimal impact on DBP
and the afnity for corresponding Tyrosine Kinase Inhibitors.
Recently, the variant-specic landscape of mutual exclusivity among
BRAF, KRAS, and EGFR mutations in cancer has been unraveled206.We
learn which oncogenic variants can co-occur in the same cancer sample
whilecertain driver events aremutually exclusive. The authors concludethat
class I BRAF(in line with another recent report206,207), Hydrolysis KRAS206,
and classical-like EGFR206 class mutations are less likely to co-occur. When
they dissected the analyses into variants, they discovered novel instances of
mutual exclusivity involving unconventional yet common oncogenic var-
iants. They showed that specic classical-like EGFR and BRAF mutations,
often the most frequent ones, are mutually exclusive in human cancer206.
Leveragingoncogenes mutual exclusivity for precision oncology:
a target discovery framework for RTK/RAS/RAF pathway
agonism
Reinforced activation of the ERKp could serve as a therapeutic strategy for
ERK-associated cancers. As previously explained, these cancers exhibit
susceptibility to elevated ERKp activity beyond conventional oncogenic
levels. Importantly, this vulnerability can be selectively targeted because
normal cells and ERK-associated cancer cells differ in their baseline ERKp
activities. Thus, reinforcing ERK activation to levels intolerable for cancer
cells may not necessarily push normal cells beyond the tolerable threshold
(see Fig. 4).
At rst glance, the primary concern withthisapproachisthepotential
undesired activation of the ERKp in non-cancerous tissues. Interestingly,
this potential adverse event is not unfamiliar in precision oncology. Patients
with BRAFV600E mutant cancers have been treated with various RAF inhi-
bitors for years, initially as single agents and later combined with MEK
inhibitors. Type I RAF inhibitors like Vemurafenib and Dabrafenib can
cause paradoxical ERKp activation in non-cancerous cells with wild-type B/
CRAF. Some patients develop benign teratomas like keratoacanthoma, but
most do not26,208,209. Adding MEK inhibitors to therapy can signicantly
reduce the occurrence of such adverse events, although MEK inhibition is
also associated with toxic effects210. Furthermore, predictive markers and
signatures can help identify patients at risk of these adverse events. On the
other hand, developing selective ERKp activators with exclusive effects on
cancer cells could mitigate this challenge from the outset.
Our proposed model recognizes three non-detrimental and two det-
rimental stages and four thresholds of ERKp activity (Fig. 4). The normal
lowoccurs when cells are at rest. The normal highoccurs when cells are in
proliferating status or are stressed due to intrinsic or extrinsic signals. ERKp
activity spatiotemporally surpassing normal highphysiological levels
enters the oncogenic window.Levels below the normal lowand above
the oncogenic windowcan be detrimental. For any potential therapy, it
will be necessary to set pathway activity exceeding the oncogenic window
and entering the detrimental stage in the target cancer cells. Ideally, the
therapy should not push pathway activity beyond normal highlevels in
normal cells, adhering to the Goldilocks principle.
This discussion will not delve into the chemistry and structural aspects
of potential therapeutics, including small molecule activators, monoclonal
antibodies, monobodies, etc. We propose three strategies to reinforce the
ERKp or ERK-independent signals for detrimental effects (Fig. 5a, b).
Perturbation 1 (P1)-global activators. Potential therapeutics activate
the ERKp through various means, such as ligand-mimetic molecules
targeting EGFR, irrespective of the specic oncogenic mechanism of the
treated cell. While not selective towards their direct targets, these ther-
apeutics can exert selective detrimental effects on cancer cells. Recently,
Xu et al. from Dengs lab have introduced and characterized the rst-in-
class small molecule KRAS agonist that activates both the wild-type and
mutant KRAS molecules and has shown selective effects towards KRAS
mutant lung cancer in-vitro and in-vivo57. Another tactic involves
directly targeting the ERK1/2 proteins. Exciting research in Sugiuras lab
and colleagues has explored ERKp agonists and lead compounds that
Detrimental
Normal high
Oncogenic
Normal low
Stages of ERK pathway activity in health and disease
Fig. 4 | Simplied schematic illustrating the stages of ERKp activity in health and
disease, and upon explored and suggested therapeutic targeting. This schematic is
oversimplied and does not acknowledge the spatiotemporal contextof ERK activity and
regulation. The gure was generated using a template provided in https://www.
slideegg.com.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 8
show selective activity against ERK-associated cancer cell lines compared
to normal cells5860. Unsupervised screens involving perturbations to
activate different activating effectors of the ERKp can be applied to nd
the most effective targets.
P2-Mutation-specic targeting. Targeting protooncogenes like
BRAFV600, EGFRL858R, or KRASG12V with mutation-specic agonists can
elevate basal ERK activity beyond the oncogenic window in cancer cells,
leading to events like apoptosis or senescence. These agonists should
ideally be selective (if they ever could be) against oncogenic variants of the
targeted- versus the wild-type proteins, aligning with Paul Ehrlichs
magic bullet concept. It deserves to be mentioned that as oncogenes, like
any other protein, have a threshold of conformational stability; it cannot
be ruled out that oncogene agonists might end up destabilizing and
further orchestrating protooncogene degradation processes before even
exerting the envisioned effects.
P3-Unsupervised target discovery and targeting aided by the
variant-specic landscape of mutual exclusivity among BRAF,
EGFR, and KRAS oncogenes in human cancer. We discussed how
induction of mutually exclusive BRAF, EGFR, and KRAS oncogenes
could be detrimental in affected cells. Nature itself is presenting these
scenarios to us. While all the hypothesis-driven approaches mentioned
above show promise for further investigation, we would like to
underscore an unbiased target discovery approach guided by the
uncovered variant-specic landscape of mutual exclusivity among
BRAF, EGFR, and KRAS oncogenes. Oncogene overdose55,mimicking
the co-expression of EGFR and KRAS oncogenes56,paradoxical
intervention61,andERK-Dependent Apoptosis60 as therapeutic models
proposed by different research groups share fundamental similarities.
Nevertheless, they vary in certain specic details. The recent unveiling
of the variant-specic landscape of mutual exclusivity among ERKp
oncogenes introduces a new layer of complexity to existing concepts.
This newfound complexity offers insights into the hyperactivation of
specic signaling pathways that certain cancer cells circumvent to
avoid most hostile conditions.
Consequently, replicating these mutually exclusive scenarios could
lead to highly effective yet personalized therapies tailored for each oncogenic
variant (BRAF, KRAS, and EGFR). Therefore, in the proposed model, the
toxic effects of two oncogenesjoint expression would be limited to specic
variants, or at least more likely in those mutually exclusive scenarios.
Consequently, in our proposed model, both target (Fig. 5b) and co-target
discovery (Fig. 6) can be a matter of precision. Considering this, our model
suggests that the initial point of target discovery and potentially resulting
therapy, for instance, for the BRAFG4 69V variant, may differ from that of
BRAFV600E. Unbiased approaches, such as bulk RNA sequencing and single-
cell RNA sequencing, can help explore differentially expressed genes and
altered pathways resulting from the induction of mutually exclusive BRAF,
EGFR, and KRAS oncogenes. Genetic screens such as CRISPR screens,
especially dual LOF and GOF screens (i.e., CRISPRi and CRISPRa), may
identify the signaling nodes that govern the response and dictate cell fate
when mutually exclusive oncogenes are co-induced. These screens are
intended to elucidate targeting approaches that phenotypically replicate the
co-induction of two synthetically lethal and mutually exclusive gene
mutations. Merely Droup-out screens like KO screens may fall short in
identifying negative regulators of the ERKp as potential signaling nodes.
Of note, this approach can ideally be tailored to each tumor. For
example, for target discovery of cancer cells with BRAFV600E, one can refer to
the variant-specic landscape of mutual exclusivity database207 and nd the
most mutually exclusive scenario with BRAFV600E, and as such, design a
model with inducible expression of KRASG12D or EGFRL858R in BRAFV600E
background. In a suppressor CRISPR screen, those genes whose activation
b
a
Fig. 5 | Three Perturbations (P1-3) to reinforce the already activated ERK
pathway activity in related cancers as a therapeutic approach. a Three suggested
approaches to target the vulnerabilities to reinforced RTK/RAS/RAF pathway
activation in affected cancers. bFramework for P3 target discovery is displayed in a
more detailed manner. Figure generated in Biorender.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 9
or inhibition will lead to the rescue of senescence or synthetic lethality would
be the governing nodes and potential targets whose antagonism (when
activation rescues) or agonism (when inhibition rescues) replicate the
synthetically lethal co-induction of these mutually exclusive oncogenes (Fig.
5b).Thisapproachwidensthetargeting possibilities and may tboththe
Goldilocks principle and the magic bullet concept. Besides, this approach
may t the concept of precision oncology.
It is crucial to emphasize that the RTK/RAS/RAF effectors, the inhi-
bitors designed to target them, and their regulators can demonstrate func-
tions and interactions independently of one another, their kinase activity, or
even the ERKp itself211213. Further investigation exploring the RTK/RAS/
RAF agonism as a therapeutic approach must address whether these
functions can be leveraged to enhance oncogenic activity beyond tolerable
levels for potential therapeutic benets or, conversely, whether they might
counteract such a strategy. Therefore, any proposed approach to exploit the
susceptibility of ERK-associated cancers to hyperactivation of these effectors
should consider this notion. If the desired detrimental effect would be ERK-
independent, the P1 might fail to address it.
Widespread and highly active oncogenic variants (gene 1), in mutually
exclusive relationships with other variants (gene 2), align with the third
approach for target discovery (Fig. 5a, b: P3). Conversely, cells harboring
oncogenes with co-occurring tendencies206 (variants of gene 1 and gene 2)
may align with P1 and P2 (Fig. 5a).
Combined targeting
Cancer targeting resembles a time loop: as monotherapies are developed,
resistance emerges, driving the search for more effective combinatorial
treatments that not only offer a stronger initial impact but may also delay the
development of further resistance. Therefore, it would be wishful thinking to
predict that P1-3 would be exempt from the rise of resistance. As such,
concurrent, sequential, or intermittent combinatorial treatments can be
explored depending on the ultimate phenotypes exerted by putative ther-
apeutics. Based on the evidence revisited in this writing, opportunities for
combinatorial treatments with chemotherapeutics can be studied, whether as
double-punch or one-two-punch with senolytics. Suppose ERK agonism
would lead to immune evading phenotypes. In that case, a one-two-punch
model can be explored to address whether, after ERK agonism, the remaining
tumor could be sensitized to immune checkpoint inhibitors. The impact of
ERK agonism on cancer stem cells and in-parallel or subsequent sensitivity of
this subpopulation to chemotherapy or stem cell-targeted therapies is also an
avenue worthy of exploration. Notably, P1-P3 hold promise to be explored in
combination with therapeutics targeting the RS tolerance.
For co-target discovery, we propose that the initial conditions and
ongoing perturbations will be pre-determined using one of the P1-P3, as we
discussed for single-target discovery (Figs. 5a, b and 6). In this context, dual
loss- and GOF screens (e.g., CRISPR screens) will prove valuable in
identifying co-targets when ERK-associated cancer cells undergo the
Fig. 6 | Suggested approach for co-target discovery to more profoundly target the vulnerabilities to reinforced hyperactivation in ERK-associated human cancer.
Figure generated in Biorender.
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 10
pre-determined ERK-activating perturbation (P1-P3). This approach aims
to uncover synthetically lethal co-perturbations (SP) with P1-3, enhancing
the detrimental effects to the levels warranting higher tness threshold191
and the emergence of tumor-suppressive drug resistance mechanisms62.In
this respect, our proposed model differs from the paradoxical intervention
model, where co-perturbations are identied within the context of
hypothesis-driven constant perturbation, such as stress-inducing agents61,62.
Consequently, we envision unbiased target discovery for both the discovery
of the ERK-activating signaling nodes that govern the detrimental effects of
excessive ERKp activation and the subsequent co-target discovery phase (P3
combined with SP, see Fig. 6). Therefore, our model may precisely recapi-
tulate the mosthostile conditions cancercells are avoiding,which are theco-
induction of specic oncogenic scenarios.
Conclusions
Despite ample evidence that cancer cells are sensitive to excessive RTK/
RAS/RAF pathway activity, this approach has not been widely explored.
Perhaps years of relentless efforts to develop efcient ERK inhibitory
treatments have established a psychological barrier within us, the scientic
community, which, if not overcome, could become a dogma.
Learning from the past, we humbly recommend that all the proposed
strategies in this writing undergo unbiased preclinical exploration without
any premature preference for a specic strategy over the others.
Despite the primary focus of this writing being exclusively on the
ERKp, the mutual exclusivity of cancer-related genetic events offers a wealth
of information regarding unexplored synthetic lethality scenarios. Har-
nessing these scenarios for therapeutic purposes could open new horiz ons in
targeting cancer-related vulnerabilities. Apart from targeting oncogenes,
this approach can also encompass events related to tumor suppressors, such
as p53, especially when cancer cells remain sensitive to restoring the related
functions. Exploring these approaches and identifying targets demands a
collaborative effort involving collective benchwork and shared intellectual
contributions.
Received: 3 November 2023; Accepted: 16 February 2024;
References
1. Ehrlich, P. The collected papers of Paul Ehrlich, including a complete
bibliography/comp. and ed. by F. Himmelweit, with the assistance of
the late M. Marquardt, under the editorial direction of Sir H. Dale.
(Pergamon Press, 1956).
2. Valent, P. et al. Paul Ehrlich (1854-1915) and his contributions to the
foundation and birth of translational medicine. J. Innate Immun. 8,
111120 (2016).
3. Schweitzer, H. Ehrlichs chemotherapya new science. Science 32,
809823 (1910).
4. Bosch, F. & Rosich, L. The contributions of Paul Ehrlich to
pharmacology: a tribute on the occasion of the centenary of his
nobel prize. Pharmacology 82, 171179 (2008).
5. Gilman, A. & Philips, F. S. The biological actions and therapeutic
applications of the B-chloroethyl amines and suldes. Science 103,
409415 (1946).
6. Gilman, A. Therapeutic applications of chemical warfare agents.
Fed. Proc. 5, 285292 (1946).
7. Goodman, L. S. & Wintrobe, M. M. Nitrogen mustard therapy; use of
methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-
chloroethyl) amine hydrochloride for Hodgkins disease,
lymphosarcoma, leukemia and certain allied and miscellaneous
disorders. J. Am. Med. Assoc. 132, 126132 (1946).
8. Amjad, M. T., Chidharla, A. & Kasi, A. Cancer chemotherapy. In:
StatPearls (2022).
9. Miller, K. D. et al. Cancer treatment and survivorship statistics, 2022.
CA.Cancer J. Clin. 72, 409436 (2022).
10. Anand, U. et al. Cancer chemotherapy and beyond: current status,
drug candidates, associated risks and progress in targeted
therapeutics. Genes Dis. 10, 13671401 (2023).
11. Langreth, R. & Waldholz, M. New era of personalized medicine:
targeting drugs for each unique genetic prole. Oncologist 4,
426427 (1999).
12. Waarts, M. R., Stonestrom, A. J., Park, Y. C. & Levine, R. L. Targeting
mutations in cancer. J. Clin. Invest. 132, e154943 (2022).
13. Blass, E. & Ott, P. A. Advances in the development of personalized
neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin.
Oncol. 18, 215229 (2021).
14. Druker, B. J. et al. Effects of a selective inhibitor of the Abl tyrosine
kinase on the growth of BcrAbl positive cells. Nat. Med. 2,
561566 (1996).
15. Ottmann, O. G. et al. A phase 2 study of imatinib in patients with
relapsed or refractory Philadelphia chromosome-positive acute
lymphoid leukemias. Blood 100, 19651971 (2002).
16. Sawyers, C. L. et al. Imatinib induces hematologic and cytogenetic
responses in patients with chronic myelogenous leukemia in
myeloid blast crisis: results of a phase II study. Blood 99,
35303539 (2002).
17. Weinstein, I. B. & Joe, A. K. Mechanisms of disease: oncogene
addictiona rationale for molecular targeting in cancer therapy. Nat.
Clin. Pract. Oncol. 3, 448457 (2006).
18. Weinstein, I. B. Cancer. Addiction to oncogenesthe Achilles heal of
cancer. Science 297,6364 (2002).
19. Greenman, C. et al. Patterns of somatic mutation in human cancer
genomes. Nature 446, 153158 (2007).
20. Sjöblom, T. et al. The consensus coding sequences of human breast
and colorectal cancers. Science 314, 268274 (2006).
21. Weinstein, I. B. Disorders in cell circuitry during multistage
carcinogenesis: the role of homeostasis. Carcinogenesis 21,
857864 (2000).
22. Weinstein, I. B. et al. Disorders in cell circuitry associated with
multistage carcinogenesis: exploitable targets for cancer prevention
and therapy. Clin. Cancer Res. 3, 26962702 (1997).
23. Weinstein, I. B. & Joe, A. Oncogene addiction. Cancer Res. 68,
307780 (2008). discussion 3080.
24. Robichaux, J. P. et al. Structure-based classication predicts drug
response in EGFR-mutant NSCLC. Nature 597, 732737 (2021).
25. Cooper, A. J., Sequist, L. V. & Lin, J. J. Third-generation EGFR and
ALK inhibitors: mechanisms of resistance and management. Nat.
Rev. Clin. Oncol. 19, 499514 (2022).
26. Dankner, M., Rose, A. A. N. N., Rajkumar, S., Siegel, P. M. & Watson,
I. R. Classifying BRAF alterations in cancer: new rational therapeutic
strategies for actionable mutations. Oncogene 37,
31833199 (2018).
27. Sharma, S. V., Fischbach, M. A., Haber, D. A. & Settleman, J.
Oncogenic shock: explaining oncogene addiction through
differential signal attenuation. Clin. Cancer Res. 12,
4392s4395s (2006).
28. Moore, A. R., Rosenberg, S. C., McCormick, F. & Malek, S. RAS-
targeted therapies: is the undruggable drugged? Nat. Rev. Drug
Discov. 19, 533552 (2020).
29. De Greve, J. & Giron, P. Targeting the tyrosine kinase inhibitor-
resistant mutant EGFR pathway in lung cancer without targeting
EGFR? Transl. Lung Cancer Res. 9,13 (2020).
30. Proietti, I. et al. Mechanisms of acquired BRAF inhibitor resistance in
melanoma: a systematic review. Cancers (Basel). 12, 2801 (2020).
31. Spagnolo, F., Ghiorzo, P. & Queirolo, P. Overcoming resistance to
BRAF inhibition in BRAF-mutated metastatic melanoma.
Oncotarget 5, 1020610221 (2014).
32. Tanaka, N. et al. Clinical acquired resistance to KRAS(G12C)
inhibition through a novel KRAS switch-II pocket mutation and
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 11
polyclonal alterations converging on RAS-MAPK reactivation.
Cancer Discov. 11, 19131922 (2021).
33. Awad, M. M. et al. Acquired resistance to KRAS(G12C) inhibition in
cancer. N. Engl. J. Med. 384, 23822393 (2021).
34. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next
generation. Cell 144, 646674 (2011).
35. Morris, L. G. T. & Chan, T. A. Therapeutic targeting of tumor
suppressor genes. Cancer 121, 13571368 (2015).
36. Hassin, O. & Oren, M. Drugging p53 in cancer: one protein, many
targets. Nat. Rev. Drug Discov.https://doi.org/10.1038/s41573-
022-00571-8 (2022).
37. Moaven, O., Mangieri, C. W., Stauffer, J. A., Anastasiadis, P. Z. &
Borad, M. J. Evolving role of oncolytic virotherapy: challenges and
prospects in clinical practice. JCO Precis. Oncol. 432441 (2021)
https://doi.org/10.1200/PO.20.00395.
38. Bressy, C., Hastie, E. & Grdzelishvili, V. Z. Combining oncolytic
virotherapy with p53 tumor suppressor gene therapy. Mol. Ther.
Oncolytics 5,2040 (2017).
39. Araki, H. et al. Oncolytic virus-mediated p53 overexpression
promotes immunogenic cell death and efcacy of PD-1 blockade in
pancreatic cancer. Mol. Ther. Oncolytics 27,313 (2022).
40. Tian, Y., Xie, D. & Yang, L. Engineering strategies to enhance
oncolytic viruses in cancer immunotherapy. Signal Transduct.
Target. Ther. 7, 117 (2022).
41. Dobzhansky, T. Genetics of natural populations; recombination and
variability in populations of Drosophila pseudoobscura. Genetics 31,
269290 (1946).
42. Kaelin, W. G. Synthetic lethality: a framework for the development of
wiser cancer therapeutics. Genome Med. 1, 99 (2009).
43. Unni, A. M., Lockwood, W. W., Zejnullahu, K., Lee-Lin, S.-Q. &
Varmus, H. Evidence that synthetic lethality underlies the mutual
exclusivity of oncogenic KRAS and EGFR mutations in lung
adenocarcinoma. Elife 4, e06907 (2015).
44. Beijersbergen, R. L., Wessels, L. F. A. & Bernards, R. Synthetic
lethality in cancer therapeutics. Annu. Rev. Cancer Biol. 1,
141161 (2017).
45. Nijman, S. M. B. Synthetic lethality: general principles, utility and
detection using genetic screens in human cells. FEBS Lett. 585,
16 (2011).
46. Tung, N. & Garber, J. E. PARP inhibition in breast cancer: progress
made and future hopes. NPJ Breast Cancer 8, 47 (2022).
47. Wang, L., Lankhorst, L. & Bernards, R. Exploiting senescence for the
treatment of cancer. Nat. Rev. Cancer 22, 340355 (2022).
48. Tan, S., Li, D. & Zhu, X. Cancer immunotherapy: pros, cons and
beyond. Biomed. Pharmacother. 124, 109821 (2020).
49. Hamdan, F. & Cerullo, V. Cancer immunotherapies: a hope for the
uncurable? Front. Mol. Med.14, 73 (2023).
50. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer
immunotherapy: from T cell basic science to clinical practice. Nat.
Rev. Immunol. 20, 651668 (2020).
51. Atkins, M. B. et al. Combination dabrafenib and trametinib versus
combination nivolumab and ipilimumab for patients with advanced
BRAF-mutant melanoma: the DREAMseq trialECOG-ACRIN
EA6134. J. Clin. Oncol.https://doi.org/10.1200/JCO.22.
01763 (2022).
52. Wang, S., Xie, K. & Liu, T. Cancer immunotherapies: from efcacy to
resistance mechanisms - not only checkpoint matters. Front.
Immunol. 12, 690112 (2021).
53. Cisowski, J., Sayin, V. I., Liu, M., Karlsson, C. & Bergo, M. O.
Oncogene-induced senescence underlies the mutual exclusive
nature of oncogenic KRAS and BRAF. Oncogene 35,
13281333 (2016).
54. Mure, E., Library, T. P. & Staff, T. P. L. The story of the three bears:
metrically related, with illustrations locating it at cecil lodge, in
September 1831. (Toronto Public Library, 2010).
55. Dipak Amin, A., Rajan, S., Groysman, M. J., Pongtornpipat, P. &
Schatz, J. H. Oncogene overdose: too much of a bad thing for
oncogene-addicted cancer cells. Biomark. Cancer 7,72 (2015).
56. Varmus, H., Unni, A. M. & Lockwood, W. W. How cancer genomics
drives cancer biology: does synthetic lethality explain mutually
exclusive oncogenic mutations? Cold Spring Harb. Symp. Quant.
Biol. 81, 247255 (2016).
57. Xu, K. et al. Small molecule KRAS agonist for mutant KRAS cancer
therapy. Mol. Cancer 18, 85 (2019).
58. Satoh, R. et al. Discovery of new benzhydrol biscarbonate esters as
potent and selective apoptosis inducers of human melanomas
bearing the activated ERK pathway: SAR studies on an ERK MAPK
signaling modulator, ACA-28. Bioorg. Chem. 103, 104137 (2020).
59. Satoh, R. et al. Identication of ACA-28, a 1-acetoxychavicol
acetate analogue compound, as a novel modulator of ERK MAPK
signaling, which preferentially kills human melanoma cells. Genes
Cells 22, 608618 (2017).
60. Sugiura, R., Satoh, R. & Takasaki, T. ERK: a double-edged sword in
cancer. ERK-dependent apoptosis as a potential therapeutic
strategy for cancer. Cells 10, 2509 (2021).
61. Dias, M. H. & Bernards, R. Playing cancer at its own game: activating
mitogenic signaling as a paradoxical intervention. Mol. Oncol. 15,
19751985 (2021).
62. Dias, M. H. et al. Paradoxical activation of oncogenic signaling as a
cancer treatment strategy. bioRxiv,https://doi.org/10.1101/2023.
02.06.527335 (2023).
63. Roskoski, R. J. The ErbB/HER family of protein-tyrosine kinases and
cancer. Pharmacol. Res. 79,3474 (2014).
64. Roskoski, R. ERK1/2 MAP kinases: Structure, function, and
regulation. Pharmacol. Res. 66, 105143 (2012).
65. Ullah, R., Yin, Q., Snell, A. H. & Wan, L. RAF-MEK-ERK pathway in
cancer evolution and treatment. Semin. Cancer Biol. 85,
123154 (2022).
66. Simanshu, D. K., Nissley, D. V. & McCormick, F. RAS proteins and
their regulators in human disease. Cell 170,1733 (2017).
67. Zarrin, A. A., Bao, K., Lupardus, P. & Vucic, D. Kinase inhibition in
autoimmunity and inammation. Nat. Rev. Drug Discov. 20,
3963 (2021).
68. Rai, S. N. et al. The Role of PI3K/Akt and ERK in neurodegenerative
disorders. Neurotox. Res. 35, 775795 (2019).
69. Verschelden, G. et al. Signicant response to dabrafenib in a patient
with ErdheimChester disease with BRAFV600E mutation. Polish.
Arch. Intern. Med. 128, 386388 (2018).
70. Wee, P. & Wang, Z. Epidermal growth factor receptor cell
proliferation signaling pathways. Cancers (Basel). 9, 52 (2017).
71. Du, Z. & Lovly, C. M. Mechanisms of receptor tyrosine kinase
activation in cancer. Mol. Cancer 17, 58 (2018).
72. Santos, E. & Nebreda, A. R. Structural and functional properties of
ras proteins. FASEB J. 3, 21512163 (1989).
73. Johnson, C., Burkhart, D. L. & Haigis, K. M. Classication of KRAS-
activating mutations and the implications for therapeutic
intervention. Cancer Discov. 12, 913923 (2022).
74. Nussinov, R., Tsai, C.-J. & Jang, H. Does ras activate Raf and PI3K
allosterically? Front. Oncol. 9, 1231 (2019).
75. Ünal, E. B., Uhlitz, F. & Blüthgen, N. A compendium of ERK targets.
FEBS Lett. 591, 26072615 (2017).
76. Lee, S., Rauch, J. & Kolch, W. Targeting MAPK signaling in cancer:
mechanisms of drug resistance and sensitivity. Int. J. Mol. Sci. 21,
1102 (2020).
77. Sinkala, M., Nkhoma, P., Mulder, N. & Martin, D. P. Integrated
molecular characterisation of the MAPK pathways in human cancers
reveals pharmacologically vulnerable mutations and gene
dependencies. Commun. Biol. 4, 9 (2021).
78. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer
genome Atlas. Cell 173, 321337.e10 (2018).
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 12
79. Smorodinsky-Atias, K., Soudah, N. & Engelberg, D. Mutations that
confer drug-resistance, oncogenicity and intrinsic activity on the
ERK MAP kinases-current state of the art. Cells 9, 129 (2020).
80. Jha, S. et al. Dissecting therapeutic resistance to ERK inhibition. Mol.
Cancer Ther. 15, 548559 (2016).
81. Jaiswal, B. S. et al. ERK mutations and amplication confer
resistance to ERK-inhibitor therapy. Clin. Cancer Res. 24,
40444055 (2018).
82. Sang, D. et al. Ancestral reconstruction reveals mechanisms of ERK
regulatory evolution. Elife 8, e38805 (2019).
83. Taylor, C. A. 4th et al. Functional divergence caused by mutations in
an energetic hotspot in ERK2. Proc. Natl Acad. Sci. USA 116,
1551415523 (2019).
84. Levin-Salomon, V., Kogan, K., Ahn, N. G., Livnah, O. & Engelberg, D.
Isolation of intrinsically active (MEK-independent) variants of the
ERK family of mitogen-activated protein (MAP) kinases. J. Biol.
Chem. 283, 3450034510 (2008).
85. Kushnir, T. et al. An activating mutation in ERK causes hyperplastic
tumors in a scribble mutant tissue in drosophila. Genetics 214,
109120 (2020).
86. Smorodinsky-Atias, K. et al. Intrinsically active variants of Erk
oncogenically transform cells and disclose unexpected
autophosphorylation capability that is independent of TEY
phosphorylation. Mol. Biol. Cell 27, 10261039 (2016).
87. Soudah, N. et al. A conserved arginine within the αC-helix of Erk1/2 is
a latch of autoactivation and of oncogenic capabilities. J. Biol.
Chem. 299, 105072 (2023).
88. Goetz, E. M., Ghandi, M., Treacy, D. J., Wagle, N. & Garraway, L. A.
ERK mutations confer resistance to mitogen-activated protein
kinase pathway inhibitors. Cancer Res. 74, 70797089 (2014).
89. Leung, G. P. et al. Hyperactivation of MAPK signalingis deleterious to
RAS/RAF-mutant melanoma. Mol. Cancer Res. 17, 199211 (2019).
90. Brenan, L. et al. Phenotypic characterization of a comprehensive set
of MAPK1/ERK2 missense mutants. Cell Rep. 17, 11711183 (2016).
91. Noeparast, A. et al. CRAF mutations in lung cancer can be
oncogenic and predict sensitivity to combined type II RAF and MEK
inhibition. Oncogene 38, 59335941 (2019).
92. Riaud, M. et al. The role of CRAF in cancer progression: from
molecular mechanisms to precision therapies. Nat. Rev. Cancer 24,
105122 (2024).
93. Emuss, V., Garnett, M., Mason, C. & Marais, R. Mutations of C-RAF
are rare in human cancer because C-RAF has a low basal kinase
activity compared with B-RAF. Cancer Res. 65, 9719 (2005).
94. Yang, L., Zheng, L., Chng, W. J. & Ding, J. L.Comprehensive analysis
of ERK1/2 substrates for potential combination immunotherapies.
Trends Pharmacol. Sci. 40, 897910 (2019).
95. Prasad, M. et al. MEK1/2inhibitiontransiently alters the tumor immune
microenvironment to enhance immunotherapy efcacy against head
and neck cancer. J. Immunother. Cancer 10, e003917 (2022).
96. Coelho, M. A. et al. Oncogenic RAS signaling promotes tumor
immunoresistance by stabilizing PD-L1 mRNA. Immunity 47,
10831099.e6 (2017).
97. Cagnol, S. & Chambard, J. C. ERK and cell death: mechanisms of
ERK-induced cell death - apoptosis, autophagy and senescence.
FEBS J. 277,221 (2010).
98. Wu, P.-K., Becker, A. & Park, J.-I. Growth inhibitory signaling of the
Raf/MEK/ERK pathway. Int. J. Mol. Sci. 21, 5436 (2020).
99. Yue, J. & López, J. M. Understanding MAPK signaling pathways in
apoptosis. Int. J. Mol. Sci. 21, 2346 (2020).
100. Yuan, Y. et al. Activation of ERK-Drp1 signaling promotes hypoxia-
induced Aβaccumulation by upregulating mitochondrial ssion and
BACE1 activity. FEBS Open Bio 11, 27402755 (2021).
101. He, K. & Aizenman, E. ERK signaling leads to mitochondrial
dysfunction in extracellular zinc-induced neurotoxicity. J.
Neurochem. 114, 452461 (2010).
102. Cook, S. J., Stuart, K., Gilley, R. & Sale, M. J. Control of cell death and
mitochondrial ssion by ERK1/2 MAP kinase signalling. FEBS J. 284,
41774195 (2017).
103. Rezatabar, S. et al. RAS/MAPK signaling functions in oxidative
stress, DNA damage response and cancer progression. J. Cell.
Physiol. 234, 1495114965 (2019).
104. Luo, Z. et al. Hypoxia signaling in human health and diseases:
implications and prospects for therapeutics. Signal Transduct.
Target. Ther. 7, 218 (2022).
105. Zhang, J. et al. ROS and ROS-mediated cellular signaling. Oxid.
Med. Cell. Longev. 2016, 4350965 (2016).
106. Papa, S., Choy, P. M. & Bubici, C. The ERK and JNK pathways in the
regulation of metabolic reprogramming. Oncogene 38,
22232240 (2019).
107. McNeal, A. S. et al. BRAF\textsuperscript{V600E} induces reversible
mitotic arrest in human melanocytes via microRNA-mediated
suppression of AURKB. Elife 10, e70385 (2021).
108. Michaloglou, C. et al. BRAFE600-associated senescence-like cell
cycle arrest of human naevi. Nature 436, 720724 (2005).
109. Hong, S.-K. K., Wu, P.-K. K. & Park, J.-I. I. A cellular threshold for
active ERK1/2 levels determines Raf/MEK/ERK-mediated growth
arrest versus death responses. Cell. Signal. 42,1120 (2018).
110. Wu, P.-K. et al. A mortalin/HSPA9-mediated switch in tumor-
suppressive signaling of Raf/MEK/extracellular signal-regulated
kinase. Mol. Cell. Biol. 33, 40514067 (2013).
111. Arthan, D., Hong, S.-K. & Park, J.-I. Leukemia inhibitory factor can
mediate Ras/Raf/MEK/ERK-induced growth inhibitory signaling in
medullary thyroid cancer cells. Cancer Lett. 297,3141 (2010).
112. Hong, S.-K., Kim, J.-H., Lin, M.-F. & Park, J.-I. The Raf/MEK/
extracellular signal-regulated kinase 1/2 pathway can mediate
growth inhibitory and differentiation signaling via androgen receptor
downregulation in prostate cancer cells. Exp. Cell Res. 317,
26712682 (2011).
113. Hong, S.-K., Yoon, S., Moelling, C., Arthan, D. & Park, J.-I.
Noncatalytic function of ERK1/2 can promote Raf/MEK/ERK-
mediated growth arrest signaling. J. Biol. Chem. 284,
3300633018 (2009).
114. Blagosklonny, M. V., Schulte, T., Nguyen, P., Trepel, J. & Neckers, L.
M. Taxol-induced apoptosis and phosphorylation of Bcl-2 protein
involves c-Raf-1 and represents a novel c-Raf-1 signal transduction
pathway. Cancer Res. 56, 18511854 (1996).
115. Xiao, D. & Singh, S. V. Phenethyl isothiocyanate-induced apoptosis
in p53-decient PC-3 human prostate cancer cell line is mediated by
extracellular signal-regulated kinases. Cancer Res. 62,
36153619 (2002).
116. Tang, D. et al. ERK activation mediates cell cycle arrest and
apoptosis after DNA damage independently of p53. J. Biol. Chem.
277, 1271012717 (2002).
117. Hsu, Y., Kuo, P., Lin, L. & Lin, C. Asiatic acid, a triterpene, induces
apoptosis and cell cycle arrest through activation of extracellular
signal-regulated kinase and p38 mitogen-activated protein kinase
pathways in human breast cancer cells. J. Pharmacol. Exp. 313,
333344 (2005).
118. Calcabrini, A. et al. Inhibition of proliferation and induction of
apoptosis in human breast cancer cells by lauryl gallate.
Carcinogenesis 27, 16991712 (2006).
119. Ciccarelli, C. et al. p21WAF1 expression induced by MEK/ERK
pathway activation or inhibition correlates with growth arrest,
myogenic differentiation and onco-phenotype reversal in
rhabdomyosarcoma cells. Mol. Cancer 4, 41 (2005).
120. Guégan, J. P., Ezan, F., Gailhouste, L., Langouët, S. & Baffet, G.
MEK1/2 overactivation can promote growth arrest by mediating
ERK1/2-dependent phosphorylation of p70S6K. J. Cell. Physiol.
229, 903915 (2014).
121. Lane, D. P. p53, guardian of the genome. Nature 358,1516 (1992).
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 13
122. Eischen, C. M. Genome stability requires p53. Cold Spring Harb.
Perspect. Med. 6, a026096 (2016).
123. Bieging, K. T., Mello, S. S. & Attardi, L. D. Unravelling mechanisms of
p53-mediated tumour suppression. Nat. Rev. Cancer 14,
35970 (2014).
124. Vaseva, A. V. et al. P53 opens the mitochondrial permeability
transition pore to trigger necrosis. Cell 149, 15361548 (2012).
125. Marchenko, N. D. & Moll, U. M. Mitochondrial death functions of p53.
Mol. Cell. Oncol. 1, e955995 (2014).
126. Timofeev, O. et al. Residual apoptotic activity of a tumorigenic p53
mutant improves cancer therapy responses. EMBO J. https://doi.
org/10.15252/embj.2019102096 (2019).
127. Jiang, P. et al. P53 regulates biosynthesis through direct inactivation
of glucose-6-phosphate dehydrogenase. Nat. Cell Biol.https://doi.
org/10.1038/ncb2172 (2011).
128. Zhao, Y. et al. p53 translocation to mitochondria precedes its nuclear
translocation and targets mitochondrial oxidative defense protein-
manganese superoxide dismutase. Cancer Res.https://doi.org/10.
1158/0008-5472.CAN-04-3835 (2005).
129. Tasdemir, E. et al. Regulation of autophagy by cytoplasmic p53. Nat.
Cell Biol. 10, 676687 (2008).
130. Hampp, S. et al. DNA damage tolerance pathway involving DNA
polymerase ιand the tumor suppressor p53 regulates DNA
replication fork progression. Proc. Natl. Acad. Sci. USA.https://doi.
org/10.1073/pnas.1605828113 (2016).
131. Linke, S. P. et al. p53 interacts with hRAD51 and hRAD54, and
directly modulates homologous recombination. Cancer Res. 63,
25962605 (2003).
132. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W.
Oncogenic ras provokes premature cell senescence associated with
accumulation of p53 and p16INK4a. Cell 88, 593602 (1997).
133. Braig, M. et al. Oncogene-induced senescence as an initial barrier in
lymphoma development. Nature 436, 660665 (2005).
134. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in
suppression of Pten-decient tumorigenesis. Nature 436,
725730 (2005).
135. Collado, M. et al. Tumour biology: senescence in premalignant
tumours. Nature 436, 642 (2005).
136. Courtois-Cox, S. et al. A negative feedback signaling network
underlies oncogene-induced senescence. Cancer Cell 10,
459472 (2006).
137. Brown, L. & Benchimol, S. The involvement of MAPK signaling
pathways in determining the cellular response to p53 activation: cell
cycle arrest or apoptosis. J. Biol. Chem. 281, 38323840 (2006).
138. Bates, S. et al. p14ARF links the tumour suppressors RB and p53.
Nature 395, 124125 (1998).
139. Persons, D. L., Yazlovitskaya, E. M. & Pelling, J. C. Effect of
extracellular signal-regulated kinase on p53 accumulation in
response to cisplatin. J. Biol. Chem. 275, 3577835785 (2000).
140. Okuno, T., Matsuoka, M., Sumizawa, T. & Igisu, H. Involvement of the
extracellular signal-regulated protein kinase pathway in
phosphorylation of p53 protein and exerting cytotoxicity in human
neuroblastoma cells (SH-SY5Y) exposed to acrylamide. Arch.
Toxicol. 80, 146153 (2006).
141. She, Q. B., Chen, N. & Dong, Z. ERKs and p38 kinase phosphorylate
p53 protein at serine 15 in response to UV radiation. J. Biol. Chem.
275, 2044420449 (2000).
142. Yeh, P. Y. et al. Phosphorylation of p53 on Thr55 by ERK2 is
necessary for doxorubicin-induced p53 activation and cell death.
Oncogene 23, 35803588 (2004).
143. Collado, M. & Serrano, M. Senescence in tumours: evidence from
mice and humans. Nat. Rev. Cancer.https://doi.org/10.1038/
nrc2772 (2010).
144. Astle, M. V. et al. AKT induces senescence in human cells via
mTORC1 and p53 in the absence of DNA damage: implications for
targeting mTOR during malignancy. Oncogene 31,
19491962 (2012).
145. Lowe, S. W., Cepero, E. & Evan, G. Intrinsic tumour suppression.
Nature 432, 30715 (2004).
146. Drosten, M. et al. Loss of p53 induces cell proliferation via Ras-
independent activation of the Raf/Mek/Erk signaling pathway. Proc.
Natl Acad. Sci. USA 111, 1515515160 (2014).
147. Karkhanis, M. & Park, J.-I. Sp1 regulates Raf/MEK/ERK-induced
p21(CIP1) transcription in TP53-mutated cancer cells. Cell Signal.
27, 479486 (2015).
148. De, S., Campbell, C., Venkitaraman, A. R. & Esposito, A. Pulsatile
MAPK Signaling Modulates p53 activity to control cell fate decisions
at the G2 checkpoint for DNA damage. Cell Rep. 30,
20832093.e5 (2020).
149. Marchetti, A. et al. p53 can inhibit cell proliferation through caspase-
mediated cleavage of ERK2/MAPK. Cell Death Differ. 11,
596607 (2004).
150. Sabapathy, K. & Lane, D. P. Therapeutic targeting of p53: all mutants
are equal, but some mutants are more equal than others. Nat. Rev.
Clin. Oncol. https://doi.org/10.1038/nrclinonc.2017.151 (2017).
151. Donehower, L. A. et al. Integrated analysis of TP53 gene and
pathway alterations in the cancer genome atlas. Cell Rep. 28,
13701384.e5 (2019).
152. Campbell, P. J. et al. Pan-cancer analysis of whole genomes. Nature.
https://doi.org/10.1038/s41586-020-1969-6 (2020).
153. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature.
https://doi.org/10.1038/s41586-019-1907-7 (2020).
154. Kandoth, C. et al. Mutational landscape and signicance across 12
major cancer types. Nature 502, 333339 (2013).
155. Leroy, B., Anderson, M. & Soussi, T. TP53 mutations in human
cancer: database reassessment and prospects for the next decade.
Hum. Mutat. https://doi.org/10.1002/humu.22552 (2014).
156. Steele, C. D. et al. Signatures of copy number alterations in human
cancer. Nature 606, 984991 (2022).
157. Rivlin, N., Brosh, R., Oren, M. & Rotter, V. Mutations in the p53 tumor
suppressor gene: important milestones at the various steps of
tumorigenesis. Genes Cancer 2, 466474 (2011).
158. Chen, X. et al. Mutant p53 in cancer: from molecular mechanism to
therapeutic modulation. Cell Death Dis. 13,114 (2022).
159. Hainaut, P. & Pfeifer, G. P. Somatic TP53 mutations in the era of
genome sequencing. Cold Spring Harb. Perspect. Med. 6,
a026179 (2016).
160. Mantovani, F., Collavin, L. & Del Sal, G. Mutant p53 as a guardian of
the cancer cell. Cell Death Differ. https://doi.org/10.1038/s41418-
018-0246-9 (2019).
161. Stiewe, T. & Haran, T. E. How mutations shape p53 interactions with
the genome to promote tumorigenesis and drug resistance. Drug
Resist. Updat. 38,2743 (2018).
162. Klimovich, B. et al. p53 partial loss-of-function mutations sensitize to
chemotherapy. Oncogene 41, 10111023 (2022).
163. Skoulidis, F. & Heymach, J. V. Co-occurring genomic alterations in
non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer 19,
495509 (2019).
164. Klimovich, B. et al. Partial p53 reactivation is sufcient to induce
cancer regression. J. Exp. Clin. Cancer Res. 41, 80 (2022).
165. Kotler, E. et al. A systematic p53 mutation library links differential
functional impact to cancer mutation pattern and evolutionary
conservation. Mol. Cell 71, 178190.e8 (2018).
166. Boettcher, S. et al. A dominant-negative effect drives selection of
TP53 missense mutations in myeloid malignancies. Science 365,
599 LP604 (2019).
167. Kato, S. et al. Understanding the functionstructure and
functionmutation relationships of p53 tumor suppressor protein by
high-resolution missense mutation analysis. Proc. Natl Acad. Sci.
100, 8424 LP8429 (2003).
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 14
168. Giacomelli, A .O. et al. Mutational pr ocesses shape the landscape of
TP53 mutations in human cancer. Nat. Genet. 50,
13811387 (2018).
169. Panagopoulos, A. & Altmeyer, M. The hammer and the dance of cell
cycle control. Trends Biochem. Sci. 46, 301314 (2021).
170. Matthews, H. K., Bertoli, C. & de Bruin, R. A. M. Cell cycle control in
cancer. Nat. Rev. Mol. Cell Biol. 23,7488 (2022).
171. Gaillard, H., García-Muse, T. & Aguilera, A. Replication stress and
cancer. Nat. Rev. Cancer 15, 276289 (2015).
172. Petropoulos, M., Champeris Tsaniras, S., Taraviras, S. & Lygerou, Z.
Replication licensing aberrations, replication stress, and genomic
instability. Trends Biochem. Sci. 44, 752764 (2019).
173. da Costa, A. A. B. A., Chowdhury, D., Shapiro, G. I., DAndrea, A. D. &
Konstantinopoulos, P. A. Targeting replication stress in cancer
therapy. Nat. Rev. Drug Discov. 22,3858 (2023).
174. Bowry, A., Kelly, R. D. W. & Petermann, E. Hypertranscription and
replication stress in cancer. Trends Cancer 7, 863877 (2021).
175. Kotsantis, P., Petermann, E. & Boulton, S. J. Mechanisms of
oncogene-induced replication stress: Jigsaw falling into place.
Cancer Discov. 8, 537555 (2018).
176. Segeren, H. A., van Liere, E. A., Riemers, F. M., de Bruin, A. &
Westendorp, B. Oncogenic RAS sensitizes cells to drug-induced
replication stress via transcriptional silencing of P53. Oncogene 41,
27192733 (2022).
177. Schauer, D. et al. Clonal dynamics of BRAF-driven drug resistance
in EGFR-mutant lung cancer. NPJ Precis. Oncol. 5, 102 (2021).
178. McCubrey, J. A. et al. Roles of the Raf/MEK/ERK pathway in cell
growth, malignant transformation and drug resistance. Biochim.
Biophys. Acta 1773, 12631284 (2007).
179. Ohashi, K. et al. Lung cancers with acquired resistance to EGFR
inhibitors occasionally harbor BRAF gene mutations but lack
mutations in KRAS, NRAS, or MEK1. Proc. Natl Acad. Sci. USA 109,
E212733 (2012).
180. Aissa, A. F. et al. Single-cell transcriptional changes associated with
drug tolerance and response to combination therapies in cancer.
Nat. Commun. 12, 1628 (2021).
181. Awad, M. M. et al. Acquired resistance to KRASG12C inhibition in
cancer. N. Engl. J. Med. 384, 23822393 (2021).
182. Xue, Y. et al. An approach to suppress the evolution of resistance in
BRAF(V600E)-mutant cancer. Nat. Med. 23, 929937 (2017).
183. Tétu, P. et al. Mitogen-activated protein kinase blockade in
melanoma: intermittent versus continuous therapy, from preclinical
to clinical data. Curr. Opin. Oncol. 33, 127132 (2021).
184. Kavran, A. J. et al. Intermittent treatment of BRAF(V600E) melanoma
cells delays resistance by adaptive resensitization to drug
rechallenge. Proc. Natl Acad. Sci. USA 119, e2113535119 (2022).
185. Kong, X. et al. Cancer drug addiction is relayed by an ERK2-
dependent phenotype switch. Nature 550, 270274 (2017).
186. Gutierrez-Prat, N. et al. DUSP4 protects BRAF- and NRAS-mutant
melanoma from oncogene overdose through modulation of MITF.
Life Sci. Alliance 5, e202101235 (2022).
187. Stagno, A. et al. Case report: rechallenge with BRAF and MEK
inhibitors in metastatic melanoma: a further therapeutic option in
salvage setting? Front. Oncol. 11, 645008 (2021).
188. Matter, A. V., Micaletto, S., Urner-Bloch, U., Dummer, R. &
Goldinger, S. M. Long-term response to intermittent binimetinib in
patients with NRAS-mutant melanoma. Oncologist 25,
e1593e1597 (2020).
189. Das Thakur, M. et al. Modelling vemurafenib resistance in melanoma
reveals a strategy to forestall drug resistance. Nature 494,
251255 (2013).
190. Hong, A. et al. Exploiting drug addiction mechanisms to select
against mapki-resistant melanoma. Cancer Discov. 8,7493 (2018).
191. Xue, Y. et al. An approach to suppress the evolution of resistance in
BRAFV600E-mutant cancer. Nat. Med. 23, 929937 (2017).
192. Holdereld, M., Deuker, M. M., McCormick, F. & McMahon, M.
Targeting RAF kinases for cancer therapy: BRAF-mutated
melanoma and beyond. Nat. Rev. Cancer 14, 455467 (2014).
193. Dooley, A. J., Gupta, A., Bhattacharyya, M. & Middleton, M. R.
Intermittent dosing with vemurafenib in BRAF V600E-mutant
melanoma: review of a case series. Ther. Adv. Med. Oncol. 6,
262266 (2014).
194. Smalley, I. et al. Leveraging transcriptional dynamics to improve
BRAF inhibitor responses in melanoma. EBioMedicine 48,
178190 (2019).
195. Schreuer, M. et al. Combination of dabrafenib plus trametinib for
BRAF and MEK inhibitor pretreated patients with advanced
BRAF(V600)-mutant melanoma: an open-label, single arm, dual-
centre, phase 2 clinical trial. Lancet Oncol. 18, 464472 (2017).
196. Valpione, S. et al. Rechallenge with BRAF-directed treatment in
metastatic melanoma: a multi-institutional retrospective study. Eur.
J. Cancer 91, 116124 (2018).
197. Tietze, J. K. et al. The efcacy of re-challenge with BRAF inhibitors
after previous progression to BRAF inhibitors in melanoma: a
retrospective multicenter study. Oncotarget 9, 3433634346 (2018).
198. Algazi, A. P. et al. Continuous versus intermittent BRAF and MEK
inhibition in patients with BRAF-mutated melanoma: a randomized
phase 2 trial. Nat. Med. 26, 15641568 (2020).
199. Gonzalez-Cao, M. et al. Intermittent BRAF inhibition in advanced
BRAF mutated melanoma results of a phase II randomized trial. Nat.
Commun. 12, 7008 (2021).
200. Petti, C. et al. Coexpression of NRASQ61R and BRAFV600E in
human melanoma cells activates senescence and increases
susceptibility to cell-mediated cytotoxicity. Cancer Res. 66,
65036511 (2006).
201. Unni, A. M. et al. Hyperactivation of ERK by multiple mechanisms is
toxic to RTK-RAS mutation-driven lung adenocarcinoma cells. Elife
7,124 (2018).
202. Ambrogio, C., Barbacid, M. & Santamaría, D. In vivo oncogenic
conict triggered by co-existing KRAS and EGFR activating
mutationsin lung adenocarcinoma. Oncogene 36,23092318 (2017).
203. Sun, C. et al. Reversible and adaptive resistance to BRAF(V600E)
inhibition in melanoma. Nature 508, 118122 (2014).
204. le Sage, C. et al. Dual direction CRISPR transcriptional regulation
screening uncovers gene networks driving drug resistance. Sci.
Rep. 7, 17693 (2017).
205. Konermann, S. et al. Genome-scale transcriptional activation by an
engineered CRISPR-Cas9 complex. Nature 517, 583588 (2015).
206. Vaeyens, F. et al. Variant-specic landscape of mutual exclusivity
among BRAF, EGFR, and KRAS oncogenes in human cancer.
medRxiv https://doi.org/10.1101/2023.10.21.23297089. (2023).
207. Zhao, Y. et al. Assessment of RAS sependency for BRAF alterations
using cancer genomic databases. JAMA Netw. Open 4,
e2035479e2035479 (2021).
208. Whittaker, S. R. et al. Combined pan-RAF and MEK inhibition
overcomes multiple resistance mechanisms to selective RAF
inhibitors. Mol. Cancer Ther.14, 2700-11 (2015).
209. Reyes, R. et al. Clinical benet from BRAF/MEK inhibition in a double
non-V600E BRAF mutant lung adenocarcinoma: a case report. Clin.
Lung Cancer 20, e219e223 (2019).
210. Garutti, M. et al. BRAF and MEK inhibitors and their toxicities: a
meta-analysis. Cancers (Basel). 15, 141 (2022).
211. Maurer, G., Tarkowski, B. & Baccarini, M. Raf kinases in cancerroles
and therapeutic opportunities. Oncogene 30, 34773488 (2011).
212. Dorard, C. et al. RAF1 contributes to cell proliferation and STAT3
activation in colorectal cancer independently of microsatellite and
KRAS status. Oncogene 42, 16491660 (2023).
213. Eggermont, C. et al. The EGFR-STYK1-FGF1 axis sustains
functional drug tolerance to EGFR inhibitors in EGFR-mutant non-
small cell lung cancer. Cell Death Dis. 13, 611 (2022).
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 15
Acknowledgements
M.N. is grateful to Dr. ErikTeugels for years of scientic discussion aboutthe
idea behind this writing and beyond.
Author contributions
M.N. conceived and conceptualized the idea for the review and the
conceptual framework and authored the manuscript with scientic
support, authoring contribution, and intellectual input of O.T., P.G., S.L.,
and M.P. All authors critically revised the text as well. M.N. and S.L.
generated gures.
Funding
Open Access funding enabled and organized by Projekt DEAL.
Competing interests
All authors,includingM.N., O.T., P.G., S.L., and M.P., declare no competing
(nancial or non-nancial) interests.
Additional information
Correspondence and requests for materials should be addressed to
Maxim Noeparast.
Reprints and permissions information is available at
http://www.nature.com/reprints
Publishersnote Springer Nature remains neutral with regard to jurisdictional
claims in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the articles Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the
articles Creative Commons licence and your intended use is not permitted
by statutory regulation or exceeds the permitted use, you will need to
obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/.
© The Author(s) 2024
https://doi.org/10.1038/s41698-024-00554-5 Review article
npj Precision Oncology | (2024) 8:70 16
... Furthermore, substantial evidence suggests that MEK/ERK pathway modules are frequently dysregulated in myeloid leukemias due to genetic and epigenetic abnormalities in cytoplasmic tyrosine kinases and inhibitory phosphatases [93][94][95]. MEK inhibitors such as CI-104055, PD9805956, and U012657 have recently been developed [95][96][97]. However, their therapeutic effect is limited when used alone, as it inhibits cell proliferation rather than exerting a direct cytotoxic effect [98,99]. ...
Article
Full-text available
The patterns and biological functions of copper homeostasis-related genes (CHRGs) in acute myeloid leukemia (AML) remain unclear. We explored the patterns and biological functions of CHRGs in AML. Using independent cohorts, including TCGA-GTEx, GSE114868, GSE37642, and clinical samples, we identified 826 common differentially expressed genes. Specifically, 12 cuproptosis-related genes (e.g., ATP7A, ATP7B) were upregulated, while 17 cuproplasia-associated genes (e.g., ATOX1, ATP7A) were downregulated in AML. We used LASSO-Cox, Kaplan-Meier, and Nomogram analyses to establish prognostic risk models, effectively stratifying patients with AML into high- and low-risk groups. Subgroup analysis revealed that high-risk patients exhibited poorer overall survival and involvement in fatty acid metabolism, apoptosis, and glycolysis. Immune infiltration analysis indicated differences in immune cell composition, with notable increases in B cells, cytotoxic T cells, and memory T cells in the low-risk group, and increased monocytes and neutrophils in the high-risk group. Single-cell sequencing analysis corroborated the expression characteristics of critical CHRGs, such as MAPK1 and ATOX1, associated with the function of T, B, and NK cells. Drug sensitivity analysis suggested potential therapeutic agents targeting copper homeostasis, including Bicalutamide and Sorafenib. PCR validation confirmed the differential expression of 4 cuproptosis-related genes (LIPT1, SLC31A1, GCSH, and PDHA1) and 9 cuproplasia-associated genes (ATOX1, CCS, CP, MAPK1, SOD1, COA6, PDK1, DBH, and PDE3B) in AML cell line. Importantly, these genes serve as potential biomarkers for patient stratification and treatment. In conclusion, we shed light on the expression patterns and biological functions of CHRGs in AML. The developed risk models provided prognostic implications for patient survival, offering valuable information on the regulatory characteristics of CHRGs and potential avenues for personalized treatment in AML.
Preprint
Full-text available
In this cross-sectional study, we report the findings of our investigation into the mutual exclusivity (ME) and co-occurrence (CO) patterns of BRAF, KRAS, and EGFR mutations in human cancer. Our analysis acknowledges previously overlooked mutational subtypes with distinct clinical implications. Creating an automated R framework, we analyzed mutation data from 64807 unique cBioPortal samples, 1570 cell lines, and 2714 unique Belgian cancer samples. Consistently, across all three datasets, we observe that co-occurrence is less likely among class I BRAF, Hydrolysis KRAS, and Classical-like EGFR mutations. Bilateral variant-assigned CO matrices uncover novel inter-class and inter-type CO and ME scenarios, encompassing conventional and atypical mutations. Besides Class I BRAF, various mutation classes exhibit diverse CO patterns, justifying the need to refine mutational classifications. We provide a variant-specific database for precision oncology showcasing ME among three actionable oncogenes. These findings may guide the discovery of novel synthetically lethal interactions for targeted cancer therapy
Article
Full-text available
More than 30% of all human cancers are driven by RAS mutations and activating KRAS mutations are present in 40% of colorectal cancer (CRC) in the two main CRC subgroups, MSS (Microsatellite Stable) and MSI (Microsatellite Instable). Studies in RAS-driven tumors have shown essential roles of the RAS effectors RAF and specifically of RAF1, which can be dependent or independent of RAF’s ability to activate the MEK/ERK module. In this study, we demonstrate that RAF1, but not its kinase activity, plays a crucial role in the proliferation of both MSI and MSS CRC cell line-derived spheroids and patient-derived organoids, and independently of KRAS mutation status. Moreover, we could define a RAF1 transcriptomic signature which includes genes that contribute to STAT3 activation, and could demonstrate that RAF1 ablation decreases STAT3 phosphorylation in all CRC spheroids tested. The genes involved in STAT3 activation as well as STAT3 targets promoting angiogenesis were also downregulated in human primary tumors expressing low levels of RAF1. These results indicate that RAF1 could be an attractive therapeutic target in both MSI and MSS CRC regardless of their KRAS status and support the development of selective RAF1 degraders rather than RAF1 inhibitors for clinical use in combination therapies.
Article
Full-text available
The use of cancer immunotherapies is not novel but has been used over the decades in the clinic. Only recently have we found the true potential of stimulating an anti-tumor response after the breakthrough of checkpoint inhibitors. Cancer immunotherapies have become the first line treatment for many malignancies at various stages. Nevertheless, the clinical results in terms of overall survival and progression free survival were not as anticipated. Majority of cancer patients do not respond to immunotherapies and the reasons differ. Hence, further improvements for cancer immunotherapies are crucially needed. In the review, we will discuss various forms of cancer immunotherapies that are being tested or already in the clinic. Moreover, we also highlight future directions to improve such therapies.
Preprint
Full-text available
Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress-response programs that counteract the inherent toxicity of such aberrant signaling. While inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of Protein Phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor suppressive resistance.
Article
Full-text available
Purpose: This meta-analysis summarizes the incidence of treatment-related adverse events (AE) of BRAFi and MEKi. Methods: A systematic search of Medline/PubMed was conducted to identify suitable articles published in English up to 31 December 2021. The primary outcomes were profiles for all-grade and grade 3 or higher treatment-related AEs, and the analysis of single side effects belonging to both categories. Results: The overall incidence of treatment-related all-grade Aes was 99% for Encorafenib (95% CI: 0.97-1.00) and 97% for Trametinib (95% CI: 0.92-0.99; I2 = 66%) and Binimetinib (95% CI: 0.94-0.99; I2 = 0%). In combined therapies, the rate was 98% for both Vemurafenib + Cobimetinib (95% CI: 0.96-0.99; I2 = 77%) and Encorafenib + Binimetinib (95% CI: 0.96-1.00). Grade 3 or higher adverse events were reported in 69% of cases for Binimetinib (95% CI: 0.50-0.84; I2 = 71%), 68% for Encorafenib (95% CI: 0.61-0.74), and 72% for Vemurafenib + Cobimetinib (95% CI: 0.65-0.79; I2 = 84%). The most common grade 1-2 AEs were pyrexia (43%) and fatigue (28%) for Dabrafenib + Trametinib and diarrhea for both Vemurafenib + Cobimetinib (52%) and Encorafenib + Binimetinib (34%). The most common AEs of grade 3 or higher were pyrexia, rash, and hypertension for Dabrafenib + Trametinib (6%), rash and hypertension for Encorafenib + Binimetinib (6%), and increased AST and ALT for Vemurafenib + Cobimetinib (10%). Conclusions: Our study provides comprehensive data on treatment-related adverse events of BRAFi and MEKi combination therapies, showing related toxicity profiles to offer a helpful tool for clinicians in the choice of therapy.
Article
Full-text available
TP53, a crucial tumor suppressor gene, is the most commonly mutated gene in human cancers. Aside from losing its tumor suppressor function, mutant p53 (mutp53) often acquires inherent, novel oncogenic functions, which is termed “gain-of-function”. Emerging evidence suggests that mutp53 is highly associated with advanced malignancies and poor prognosis, which makes it a target for development of novel cancer therapies. Herein, we provide a summary of our knowledge of the mutp53 types and mutp53 spectrum in cancers. The mechanisms of mutp53 accumulation and gain-of-function are also summarized. Furthermore, we discuss the gain-of-function of mutp53 in cancers: genetic instability, ferroptosis, microenvironment, and stemness. Importantly, the role of mutp53 in the clinic is also discussed, particularly with regard to chemotherapy and radiotherapy. Last, emphasis is given to emerging strategies on how to target mutp53 for tumor therapy. Thus, this review will contribute to better understanding of the significance of mutp53 as a target for therapeutic strategies.
Article
The RAF family of kinases includes key activators of the pro-tumourigenic mitogen-activated protein kinase pathway. Hyperactivation of RAF proteins, particularly BRAF and CRAF, drives tumour progression and drug resistance in many types of cancer. Although BRAF is the most studied RAF protein, partially owing to its high mutation incidence in melanoma, the role of CRAF in tumourigenesis and drug resistance is becoming increasingly clinically relevant. Here, we summarize the main known regulatory mechanisms and gene alterations that contribute to CRAF activity, highlighting the different oncogenic roles of CRAF, and categorize RAF1 (CRAF) mutations according to the effect on kinase activity. Additionally, we emphasize the effect that CRAF alterations may have on drug resistance and how precision therapies could effectively target CRAF-dependent tumours. Here, we discuss preclinical and clinical findings that may lead to improved treatments for all types of oncogenic RAF1 alterations in cancer.
Article
Eukaryotic protein kinases (EPKs) adopt an active conformation following phosphorylation of a particular activation loop residue. Most EPKs spontaneously autophosphorylate this residue. While structure-function relationships of the active conformation are essentially understood, those of the 'prone-to-autophosphorylate' conformation are unclear. Here, we propose that a specific site within the αC-helix of EPKs, occupied by Arg in the MAPK Erk1/2 (Arg84/65), impacts spontaneous autophosphorylation capability. MAPKs lack spontaneous autoactivation, but we found that converting Erk1/2's Arg84/65 to various residues is sufficient to enable spontaneous autophosphorylation. Additionally, Erk1 molecules mutated in Arg84 are oncogenic. Arg84/65 thus obstructs the adoption of the 'prone-to-autophosphorylate' conformation. All MAPKs, with no exception, harbor an Arg that is equivalent to Erks' Arg84/65, while Arg is rarely found at the equivalent position in other EPKs. Furthermore, we observed that Erk1/2's Arg84/65 interacts with the DFG motif, suggesting that autophosphorylation may be inhibited by the Arg84/65-DFG interactions. Erk1/2s mutated in Arg84/65 autophosphorylate not only the TEY motif, known as critical for catalysis, but also on another residue, Thr207/188. Our MS/MS analysis revealed that a large proportion of the Erk2R65H population is phosphorylated on Thr188 or on Tyr185+Thr188 and a small fraction is phosphorylated on the TEY motif. No molecules phosphorylated on Thr183+Thr188 were detected. Thus, phosphorylation of Thr183 and Thr188 is mutually exclusive raising the possibility that not only TEY-phosphorylated molecules are active, but also those phosphorylated on Tyr185+Thr188. The effect of mutating Arg84/65 may mimic a physiological scenario in which allosteric effectors cause Erk1/2 activation by autophosphorylation.
Article
Mutations in the TP53 tumour suppressor gene are very frequent in cancer, and attempts to restore the functionality of p53 in tumours as a therapeutic strategy began decades ago. However, very few of these drug development programmes have reached late-stage clinical trials, and no p53-based therapeutics have been approved in the USA or Europe so far. This is probably because, as a nuclear transcription factor, p53 does not possess typical drug target features and has therefore long been considered undruggable. Nevertheless, several promising approaches towards p53-based therapy have emerged in recent years, including improved versions of earlier strategies and novel approaches to make undruggable targets druggable. Small molecules that can either protect p53 from its negative regulators or restore the functionality of mutant p53 proteins are gaining interest, and drugs tailored to specific types of p53 mutants are emerging. In parallel, there is renewed interest in gene therapy strategies and p53-based immunotherapy approaches. However, major concerns still remain to be addressed. This Review re-evaluates the efforts made towards targeting p53-dysfunctional cancers, and discusses the challenges encountered during clinical development.
Article
Replication stress is a major cause of genomic instability and a crucial vulnerability of cancer cells. This vulnerability can be therapeutically targeted by inhibiting kinases that coordinate the DNA damage response with cell cycle control, including ATR, CHK1, WEE1 and MYT1 checkpoint kinases. In addition, inhibiting the DNA damage response releases DNA fragments into the cytoplasm, eliciting an innate immune response. Therefore, several ATR, CHK1, WEE1 and MYT1 inhibitors are undergoing clinical evaluation as monotherapies or in combination with chemotherapy, poly[ADP-ribose]polymerase (PARP) inhibitors, or immune checkpoint inhibitors to capitalize on high replication stress, overcome therapeutic resistance and promote effective antitumour immunity. Here, we review current and emerging approaches for targeting replication stress in cancer, from preclinical and biomarker development to clinical trial evaluation. Replication stress is a cause of genome instability in cancer cells. This Review discusses strategies to increase replicative stress by inhibiting the checkpoint kinases that coordinate DNA damage response and cell cycle, as well as combination strategies with other targeted therapies.