You are on page 1of 11

Review Article

The MAPK Pathway Across Different


Malignancies: A New Perspective
Mauricio Burotto, MD; Victoria L. Chiou, MD1; Jung-Min Lee, MD1; and
Elise C. Kohn, MD1,2

The mitogen-activated protein kinase/extracellular signal-regulated (MAPK/ERK) pathway is activated by upstream genomic events
and/or activation of multiple signaling events in which information coalesces at this important nodal pathway point. This pathway is
tightly regulated under normal conditions by phosphatases and bidirectional communication with other pathways, like the protein ki-
nase B/mammalian target of rapamycin (AKT/m-TOR) pathway. Recent evidence indicates that the MAPK/ERK signaling node can
function as a tumor suppressor as well as the more common pro-oncogenic signal. The effect that predominates depends on the in-
tensity of the signal and the context or tissue in which the signal is aberrantly activated. Genomic profiling of tumors has revealed
common mutations in MAPK/ERK pathway components, such as v-raf murine sarcoma viral oncogene homolog B1 (BRAF). Currently
approved for the treatment of melanoma, inhibitors of BRAF kinase are being studied alone and in combination with inhibitors of the
MAPK and other pathways to optimize the treatment of many tumor types. Therapies targeted toward MAPK/ERK components have
various response rates when used in different solid tumors, such as colorectal cancer and ovarian cancer. Understanding the differen-
tial nature of activation of the MAPK/ERK pathway in each tumor type is critical in developing single and combination regimens,
because different tumors have unique mechanisms of primary and secondary signaling and subsequent sensitivity to drugs. Cancer
2014;000:000-000. V C 2014 American Cancer Society.

KEYWORDS: mitogen-activated protein kinase (MAPK), v-raf murine sarcoma viral oncogene homolog (BRAF), extracellular signal-
regulated kinase (ERK), mitogen-activated protein kinase-kinase (MEK), signaling, melanoma, ovarian cancer, colorectal cancer.

INTRODUCTION
Great advances have been made toward understanding the genomic characterization of solid and hematologic malignan-
cies. Collaborative projects, such as The Cancer Genome Atlas (TCGA),1 have revealed the molecular complexity of
tumors and also the challenges inherent in data interpretation and clinical application. Differentiation of targetable driver
mutations from genetic and genomic noise and passenger mutations is 1 of the most important goals of genome and epige-
nome analysis.2,3 Driver mutations lead to dysregulation of signaling pathways, increasing malignant behavior.4 It has
been demonstrated that many of these gain-of-function driver mutations are druggable, leading to the development of
small molecules and antibodies that target specific events, such as the ligand-binding site of the receptor or the adenosine
triphosphate (ATP) binding site in the kinase domain of specific kinase proteins.
The mitogen-activated protein kinase (MAPK) cascade is a critical pathway for human cancer cell survival, dissemi-
nation, and resistance to drug therapy.5 The MAPK/extracellular signal-regulated kinase (ERK) pathway is a convergent
signaling node that receives input from numerous stimuli, including internal metabolic stress and DNA damage pathways
and altered protein concentrations, as well as through signaling from external growth factors, cell-matrix interactions, and
communication from other cells.6 Mutated genes responsible for the regulation of cell fate, genome integrity, and survival
can lead to increased protein amplification and alter the tumor microenvironment, thus overactivating the pathway.7
These mutations can occur upstream in membrane receptor genes, such as epithelial growth factor receptor (EGFR),8 in
signal transducers (RAS),9 regulatory partners (Sprouty),10 and downstream kinases that belong to the MAPK/ERK path-
way itself (BRAF) (Fig. 1).11,12 Several mutations involving the MAPK/ERK pathway have been identified in human can-
cers and are ripe for targeting. Current and future drug-development efforts will need to alter and regulate tumor
signaling in this complex network of codependent pathways.

Corresponding author: Mauricio Burotto, MD, Building 10, Room 12N226, 9000 Rockville Pike, Bethesda, MD 20892; Fax: (301) 451-0456;
mauricio.burottopichun@nih.gov
1
Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; 2Cancer Therapy Evaluation Program, Division of
Cancer Treatment and Diagnostics, National Cancer Institute, Bethesda, Maryland

DOI: 10.1002/cncr.28864, Received: February 13, 2014; Revised: April 22, 2014; Accepted: May 8, 2014, Published online Month 00, 2014 in Wiley Online
Library (wileyonlinelibrary.com)

Cancer Month 00, 2014 1


Review Article

The MAPK Pathway and its Regulation Suppression of ERK activation occurred in the first hour
There are 4 independent MAPK pathways composed of 4 of treatment, in contrast to the sustained ERK phospho-
signaling families: the MAPK/ERK family or classical rylation after 9 days of treatment.24 Indeed, cells interpret
pathway and the Big MAP kinase-1 (BMK-1), c-Jun N- and respond differently to small changes in the levels of
terminal kinase (JNK), and p38 signaling families.13 MAPK/ERK activation. As described by Murphy et al, c-
These families share a basic organization composed of 2 FOS, an early gene product of MAPK/ERK activation,
serine/threonine kinases and 1 double-specificity threo- works as a sensor of the duration of ERK stimulation.
nine/tyrosine kinase.14 Generically, these kinases are des- When the MAPK/ERK signal is transient, c-FOS is unsta-
ignated from upstream to downstream, closer to the ble and degraded in the nucleus; but, if the signal is sus-
nucleus, as MAPK kinase-kinase (MAPKKK), MAPK ki- tained, then c-FOS is phosphorylated, and specific
nase (MAPKK) and MAPK (Fig. 1).5 The canonical domains are exposed, promoting more ERK activation.25
MAPK/ERK pathway is composed of 3 types of The procarcinogenic or proapoptotic signaling of this
MAPKKK: A-RAF, B-RAF, and RAF-1 or C-RAF ki- pathway depends on the timing and duration of MAPK/
nases. BRAF is the gene most commonly mutated at this ERK activation.
level in human cancer. One level below are the MAPKKs, Specific proteins, such as kinase suppressor Ras-1
which are composed of MEK1 and MEK2. Finally, fur- (KSR1), work as the main scaffold for proteins related to
ther downstream are ERK1 and ERK2, which are the final MAPK/ERK pathway activation. Cytoplasmic proteins,
effectors of the MAPK pathway.15 Sprouty and Spred, directly inhibit the pathway26 by
ERK phosphorylation results in the activation of removing activating phosphate groups from ERK, thereby
multiple substrates that are responsible for the stimulation decreasing its ability to phosphorylate its substrates.12
of cell proliferation. Spatial localization of ERK deter- Thus, there are regulatory events in both the cytoplasm
mines target substrates and later effects within the cell.6 and the nucleus, along with spatial and temporal regula-
When located in the cytoplasm, ERK phosphorylates tion, that fine tune the output of the MAPK/ERK
cytoskeletal proteins that affect cell movement and traf- pathway.
ficking,16 metabolism, cell adhesion, and nodal regulation
of other pathways.17 Cytoplasmic substrates include ribo- Overactivated and Oncogenic Drivers of the
somal S6 kinases (RSKs), which regulate the glycogen syn- MAPK Pathway as Therapeutic Targets
thase kinase 3 (GSK3) involved in metabolism, and L1 Cellular proliferation is driven by an intricate network of
adhesion molecule, a protein of neural origin that partici- regulated, interdependent signals. The complexity of the
pates in cell adhesion.18,19 Minutes after MAPK/ERK MAPK pathway is not random; it allows for the periodic
activation, ERK detaches from cytoplasmic anchoring environmental adaptation necessary for activation and
proteins and translocates to the nucleus to exert its tran- regulation of the coordinated events critical for cell sur-
scriptional regulation.20 Active ERK in the nucleus causes vival.27 MAPK/ERK pathway activation and subsequent
phosphorylation and activation of various transcription interactions are highly regulated processes that are deregu-
factors, such as carbamoyl phosphate synthetase II (CPS lated in cancer cells. Stimulation of growth factor recep-
II) linking with synthesis of DNA or p90RSK and pro- tors in the cell membrane leads to activation of 2 different
moting cell cycle progression. These 2 events are integral but interconnected, pivotal pathways: the phosphoinosi-
in MEK/ERK stimulation of cell proliferation.21,22 In tide 3-kinase (PI3K) signal, which causes activation of
immune cells, activated ERK is also a component of the protein kinase B (AKT) and its downstream substrates,
innate response in different steps of the inflammatory cas- and the MAPK/ERK pathway (Fig. 1). Both drive cell
cade, increasing the expression of tumor necrosis factor proliferation, survival, and dissemination. The PI3K/
alpha (TNF-a) and inducible nitric oxide synthase AKT pathway also promotes anabolism; whereas the
(iNOS).23 MAPK/ERK pathway is more active in proliferation and
In addition to spatial activation, the final effect of invasion.5 Up-regulation of MAPK/ERK signaling occurs
the MAPK/ERK pathway is modulated by timing, dura- as a result of overexpression or aberrant activation of re-
tion, and intensity of its signal. Winters et al examined the ceptor tyrosine kinases (RTKs) or their immediate down-
MAPK/ERK cascade at different times points in colo- stream targets: PI3K, proto-oncogene tyrosine-protein
rectal cancer (CRC) cell lines under the combination of kinase Src (SRC), and RAS.
carboxyamido-triazole, an intracellular calcium regulator, Normal MAPK/ERK function is also responsible for
plus the selective cyclooxygenase 2 inhibitor celecoxib. tumor suppression through the induction of senescence

2 Cancer Month 00, 2014


Tissue-Specific MAPK Signaling/Burotto et al

Figure 1. This is a model of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway. Af-
ter membrane receptor activation, adaptor proteins recruit RAS proteins to activate steps that conclude with ERK activation.
Successive steps of phosphorylation amplify the signal, Raf!MAPK kinase (MEK)!ERK, until ERK activates its cytoplasmic and/
or nuclear targets. Regulatory phosphatases, Sprouty and Spred, modulate the intensity of the signal. The phosphatidylinositol-3-
kinase/protein kinase B (PI3K/AKT) pathway interacts with the MAPK/ERK node under normal conditions and in the cancer cell.
Target cytoplasmic proteins include ribosomal S6 kinases (RSKs), glycogen synthase kinase 3 (GSK3), and adhesion molecule L1
(LI). Additional proteins in the nucleus include copalyl diphosphate synthase 2 (CPS2) and 90-kDa ribosomal protein S6 kinase
(p90Rsk). EGFR indicates epidermal growth factor receptor; PDGFR, platelet-derived growth factor receptor; VEGFR, vascular
endothelial growth factor receptor; RTK, receptor tyrosine kinase; SRC, proto-oncogene tyrosine-protein kinase Src; K-Ras, v-Ki-
ras Kirsten rat sarcoma viral oncogene homolog; MAPKKK, MAPK kinase-kinase; A-Raf, serine/threonine-protein kinase A-Raf;
MAPKK, MAPK kinase; mTOR, mammalian target of rapamycin.

and the ubiquitinization and degradation of proteins nec- demonstrating that MEK1 has tumor suppressor proper-
essary for cell cycle activity and survival.28 Senescence ties and that the function of MEK1 kinase is context-
involves the inhibition of cell proliferation through termi- dependent.30
nal cell cycle arrest.29 Abnormal activation of MAPK/ Genetic mutations can dysregulate kinase activity
ERK by RAS causes degradation of proteins required for and hyperactivate the MAPK pathway during induction
both migration and progression through the cell cycle, as and progression of tumorigenesis. Many oncogenic driver
demonstrated in a model of normal fibroblasts and vali- mutations have been identified in genes upstream of
dated in benign prostate tumors. In those tumors, high MAPK/ERK, varying across cancer types, as indicated in
levels of phosphorylated (phospho)-ERK were coex- Table 1. These may include exon 21 mutations in EGFR
pressed with markers of senescent p16INK4a and promy- or del19EGFR, mutations in v-Ki-ras Kirsten rat sarcoma
elocytic leukemia protein (PML), a marker of protein viral oncogene homolog (KRAS), and the classic valine to
degradation.28 In addition, a screening study using a panel glutamic acid (V600E) BRAF mutation. These mutated
of silencing RNAs (shRNAs) against MEK1 increased genes lead to downstream overactivation of the MAPK/
lymphoma genesis in MYC-expressing lymphoid cells, ERK pathway. In general, mutations affecting the

Cancer Month 00, 2014 3


Review Article

TABLE 1. Frequency of Mutations in the Activators and Components of Mitogen-Activated Protein Kinase/
Extracellular Signal-Regulated Kinase Pathway Across Different Tumors

Frequency of Mutations, % (References)

Tumor RAS BRAF MEK ERK

Melanoma 15-29 (Colombino 2012,31 50-60 (Davies 200233) 3-8 (Murugan 2009,34 NR
Edlundh-Rose 200632) Nikolaev 201235)
NSCLC 12-30 (Seo 201236) 37
4 (Cardarella 2013 ) NR NR
Colorectal 34.1 (Cancer Genome Atlas 5-20 (Davies 2002,33 Tol 200939) <3 (Murugan 200934) NR
Network 201238)
Ovarian
HGSOC 0-12 (Sieben 200440) NR NR NR
LGSOC 27-36 (Bell 2005,41 Singer 200342) 33-50 (Bell 2005,41 Singer 200342)
Thyroid 9-27 (Bansal 201343) 45-69 (Xing 2013,44 Cohen 200345) NR NR
Hairy cell NR 79-100 (Xi 2012,46 Tiacci 201147) NR NR

Abbreviations: BRAF, v-raf murine sarcoma viral oncogene homolog; ERK, extracellular signal-regulated kinase; HGSOC, high-grade serous ovarian cancer;
LGSOC, low-grade serous ovarian cancer; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; NR, not reported; NSCLC,
nonsmall cell lung cancer; RAS, v-ras viral oncogene homolog.

MAPK/ERK pathway are singular, independent events. exhibited some activity in metastatic biliary cancers in a
Infrequently, 2 mutations can be identified in the RAS/ study of 28 patients, yielding a response rate of 12% and a
RAF/MEK/ERK pathway within the same tumor, dem- progression-free survival of 3.7 months. Only 2 patients
onstrating tumoral molecular heterogeneity.48 The sensi- had RAS mutations, and neither responded to therapy.54
tivity of mutation detection depends on the dominant Selumetinib was also studied in 24 patients who had met-
population of cells represented in the tumor sample that is astatic papillary or poorly differentiated thyroid cancer
tested and, thus, may not be illustrative of the tumor as a that was refractory to radioiodine treatment. Selumetinib
whole.49 increased the iodine-124 uptake in 12 patients, and 5 of 8
Discovery of specific oncogene mutations that acti- patients had responses to radioiodine. Mutations were
vate the MAPK pathway has spurred the development of detected in 7 of the 8 patient treated; responses were
targeted therapies that apply to multiple tumor types. reported in 4 patients who had neuroblastoma RAS viral
Studies in tumor cells with mutant V600EBRAF have oncogene homolog (NRAS) mutations and in 1 patient
demonstrated that RAF kinase inhibitors prevent ERK who had a BRAF mutation.55 Docetaxel with or without
signaling.50 The selective MEK inhibitor PD0325901 selumetinib was studied in a randomized phase 2 trial of
decreased cyclin D1 protein expression, thereby decreas- 87 patients with metastatic non-small cell lung cancer
ing cell proliferation in BRAF-mutant melanoma xeno- who had KRAS mutations. The response rate was 37% in
graft models.51 High plasma concentrations of the RAF the experimental arm versus 0% in the arm without selu-
inhibitor vemurafenib are associated with strong ERK metinib (P<.0001), and the progression-free survival was
pathway inhibition. Patients with advanced-stage, 5.3 months versus 2.1 months, respectively (P5.014).56
V600EBRAF-mutated, metastatic melanoma receiving Currently, many ongoing phase 2 clinical trials are explor-
vemurafenib treatment who achieved >80% inhibition of ing the use of agents targeting BRAF (Table 2) and MEK
cytoplasmic ERK phosphorylation, as observed in paired kinases (Table 3). Many of those trials apply mutational
pretreatment and on-treatment biopsy samples, demon- analyses for study eligibility to enrich for patients who
strated clinical evidence of partial remission.52 Recently, may be most likely to benefit. Data suggest that this path-
the immunomodulatory effects of BRAF inhibition were way behavior is not consistent in all settings, which makes
examined, and it was demonstrated that these effects targeting the addictive oncogenic pathway a challenge
explained part of the efficacy of vemurafenib in mela- across different tumor types.
noma. BRAF and MEK inhibition increased the expres- The MAPK/ERK pathway is a double-edged sword.
sion of melanoma antigens in melanoma cell lines. This Generally, therapeutic inhibition of elements within this
could increase T-cell recognition of the tumor, leading to pathway has yielded some benefit. However, small mole-
a successful immunotherapeutic approach.53 cule inhibitor therapy aimed at specific protein targets
The next proteins downstream, MEK1 and MEK2, within the MAPK/ERK pathway has resulted in the devel-
have now been successfully targeted. Selumetinib opment of secondary malignancies. RAF inhibitors, as a

4 Cancer Month 00, 2014


Tissue-Specific MAPK Signaling/Burotto et al

TABLE 2. Phase 2 Ongoing Clinical Trials for BRAF MAPK/ERK pathway (Fig. 2). Thus, the relative impor-
Inhibitors in Patients With Solid Tumors tance of MAPK/ERK in cancer cells depends on the cell
Clinical Trial and/or tissue of origin, the magnitude of addictive de-
pendence on the pathway, and the mechanisms of escape
BRAF Inhibitor Melanoma Others Tumor Types
or alternative signaling.
Vemurafenib  NCT01495988  NCT01709292: Thyroid,
locally advanced Melanoma
 NCT01813214  NCT01286753:Papillary
thyroid The success of RAF kinase inhibition was a turning point
 NCT01611675  NCT01524978: Any in the treatment of melanoma. However, as with other
BRAFV600-mutant tumor
 NCT01942993  NCT01771458: Any tumor
agents, resistance to treatment occurred and was mapped
 NCT01638676 to the MAPK/ERK pathway. Primary resistance to
 NCT01781026
 NCT01586195
vemurafenib in V600EBRAF melanomas can occur
 NCT00949702 through increased cellular proliferation in response to loss
 NCT01378975 of function of tumor suppressors or dysregulation of
 NCT01248936
Dabrafenib  NCT01682213  NCT01340846: Any mechanisms that prevent apoptosis. Phosphatase and ten-
BRAFV600-mutant tumor sin homolog (PTEN) deficiency is a major mechanism
 NCT01721603  NCT01723202: Thyroid
 NCT01266967 through which the prosurvival AKT signaling pathway
 NCT01153763  NCT01336634: NSCLC becomes constitutively activated. This was observed in
with BRAF mutation
melanoma cell lines treated with vemurafenib. This
Abbreviations: BRAF, BRAF, v-raf murine sarcoma viral oncogene homolog; PTEN deficiency was accompanied by loss of induction
NCT, National Clinical Trials identifier; V600, valine mutation at BRAF codon of the proapoptotic BIM/BCL2L11 protein and resulted
400.
in primary resistance in these cell lines.71 Selective cyto-
plasmic redistribution of the transcription factor
class, may cause abnormal skin cell proliferation, leading FOXO3a led to decreased transcription of proapoptotic
to keratoacanthomas or squamous cell cancers in approxi- proteins.85,86 These findings, coupled with the reality that
mately 10% to 20% of patients.57-59 The development of not every mutation-positive tumor will respond to B-RAF
these lesions is caused by paradoxical activation of the nor- inhibitor therapy despite activating mutations in BRAF,
mal MAPK/ERK pathway in the genomically normal skin underscores the need for future research into mechanisms
keratinocytes.59 The combination of BRAF and MEK of primary resistance.
inhibitors in metastatic melanoma resulted in improved It has been observed that tumors with oncogenic
treatment safety by counterbalancing activation of the driver mutations in the MAPK/ERK pathway progress de-
normal MAPK/ERK pathway, yielding a marked reduc- spite an initial response to targeted intervention (second-
tion in the frequency of the paradoxical oncogenic skin ary resistance). Multiple secondary mechanisms of
changes.60,61 The combination of dabrafenib (a BRAF ki- resistance have been identified in melanoma, including
nase inhibitor) with trametinib (an MEK inhibitor) new activating mutations in MAPK/ERK pathway genes87
caused keratoacanthomas in 7% of patients along with and NRAS,73 increased dimers of splice variants of wild-
rare squamous cell skin cancers compared with a fre- type BRAF,50,74,88 amplification of wild-type BRAF and
quency of 19% for dabrafenib alone.60 This safer combi- MEK,72 and increased CRAF.89 New studies have demon-
nation is now under evaluation in numerous other strated that C-RAF activates the MAPK/ERK pathway
cancers. through the acquisition of secondary mutations that
increase its half-life, avoiding degradation and allowing
MAPK/ERK Pathway Susceptibility Varies by heterodimerization with B-RAF.90 Many of these mecha-
Tumor Type nisms result in paradoxical hyperactivation of ERK.13
Different morphomolecular human tumors have demon- Intratumoral heterogeneity allows multiple mechanisms
strated unexpected differential responses to signal inter- to be identified within a single patient’s tumor, such as
ruption and may develop unique mechanisms of primary unique mutations in NRAS and an alternative splice vari-
and secondary resistance (Table 4).82-84 CRC and low- ant of BRAF.91 These findings support the concept that
grade serous ovarian cancers harbor the same mutations tumors demonstrate clonal evolution and plasticity over
that are observed in melanoma, KRAS and BRAF, but time, adapting to microenvironment and pharmacologic
have very different responses to inhibition of the exposures.92

Cancer Month 00, 2014 5


Review Article

TABLE 3. Phase 2 Trials of Mitogen-Activated predisposed to susceptibility to inhibitor therapy like


Protein Kinase Inhibitors Across Tumor Types patients with melanoma. It is now routine clinical practice
Clinical Trial to test for KRAS mutation before the initiation of EGFR
inhibitors. Thus, rather than functioning as therapeutic
MAPK Inhibitor Melanoma Other Tumor Types
targets in CRC, these genomic events in the MAPK/ERK
Selumetinib  NCT01143402  NCT00888134: Any tumor pathway are validated negative predictive biomarkers for
type EGFR inhibitor intervention.
 NCT00866177  NCT00553332: Biliary
 NCT01160718: Breast CRC resistance to B-RAF inhibition has been attrib-
 NCT00780676: Breast uted to differential activation of EGFR in the cell mem-
 NCT00514761: Colorectal
 NCT01011933: Endometrial
brane, reinforcing the differential relevance of EGFR
 NCT01089101: Glioma expression across tumor types. Treatment of BRAF-mu-
 NCT01752569: Kaposi
sarcoma
tant CRC cell lines with vemurafenib resulted in a strong
 NCT00604721: Liver increase in 1068Y-EGFR phosphorylation and receptor
 NCT00372788: NSCLC activation through inhibition of cell division cycle 25C
 NCT01306045: NSCLC,
thymic (CDC25C) phosphatase, which regulates 1068Y-EGFR
 NCT00372944: Pancreatic phosphorylation. Blockade of MAPK/ERK by BRAF or
 NCT00551070: Ovarian,
peritoneum MEK inhibitors prevented CDC25C activation, resulting
 NCT00559949: Papillary in increased 1068Y-EGFR and subsequent activation of
thyroid
 NCT01843062: Thyroid
other downstream pathways, such AKT. EGFR suppres-
Trametinib  NCT01978236  NCT01827384: Any tumor sion combined with vemurafenib markedly inhibited pro-
type
 NCT01037127  NCT01943864: Biliary
liferation in CRC cells and may be a mechanism to
 NCT01328106  NCT01553851: Oral increase clinical activity.76
squamous Cross-communication between the MAPK/ERK
BRAF inhibitor  LCCC 1128:  NCT01723202: Thyroid
plus trametinib NCT01726738 pathway and parallel pathways, such as the PI3K/AKT
 NCT01072175  NCT01750918: Colorectal and wingless-type/calcium (Wnt-Ca21) pathways, is criti-
 NCT01972347
 NCT01928940 cal to abnormal proliferation and therapy resistance.
 NCT01619774 These parallel pathways are activated when the MAPK/
 NCT01978236
 NCT01915602:
ERK pathway is attenuated, and they drive cellular prolif-
Hepatocellular eration. The inhibition of PI3K or AKT or the use of
Pimasertib  NCT01693068  NCT01936363L Ovarian
Refametinib —  NCT01915589:
hypomethylating agents that secondarily block AKT sig-
Hepatocellular naling can overcome this mechanism of resistance in
 NCT01915602: vitro.78 Understanding of mechanisms of induction of
Hepatocellular
parallel signaling is needed to guide development of com-
Abbreviations: BRAF, v-raf murine sarcoma viral oncogene homolog; bination therapies. Recently, Spreafico et al demonstrated
MAPK, mitogen-activated protein kinase; NCT, National Clinical Trials iden-
tifier; NSCLC, nonsmall cell lung cancer.
a potential role of the noncanonical Wnt/Ca21 signaling
pathway in overcoming resistance of CRC to MEK inhib-
itors using cyclosporine (a Wnt/Ca11 modulator) in a
CRC model of patient-derived tumor xenografts.79 Those mod-
Targeted kinase inhibitors (TKIs) that have been benefi- els demonstrated that drug combinations blocking both a
cial in melanoma have not yielded similar activity in targeted pathway and its associated counter-regulatory
patients with CRC. RAF signaling is downstream of RAS signal can effectively abrogate resistance of CRC to BRAF
in the MAPK/ERK pathway, such that the presence of or MEK inhibitors.
BRAF and KRAS mutations in CRC should lead to sensi-
tivity to RAF-targeted agents and circumvent the inhibi- Ovarian cancer
tion of upstream signals, such as those emanating from Ovarian cancers can be classified into distinct types, some
receptor kinases. Consistent with the latter expectation, of which are characterized by genetic mutations that may
investigators demonstrated that BRAF and KRAS muta- involve the MAPK/ERK pathway. Type II ovarian cancers
tions were negative predictors of deriving a benefit from include high-grade serous tumors, and defects in DNA
cetuximab and panitumumab (EGFR inhibitor therapy) repair through the loss of normal p53 regulation are
in phase 3 clinical trials.93,94 Those patients were not observed in almost all tumors.95,96 Type I ovarian tumors

6 Cancer Month 00, 2014


Tissue-Specific MAPK Signaling/Burotto et al

TABLE 4. Mechanisms of Primary and Secondary Resistance to Tyrosine Kinase Inhibitors in Different Tumor
Types

Mechanism of Resistance (References)

Tumor Primary Secondary

Lung MET amplification (Turke 201062), BIM polymor- T790M mutation (Kobayashi 200565), EGFR ampli-
phism (Ng 201263 and Nakagawa 201364) fication (Sequist 201166), Her2 amplification
(Takezawa 201267), PIK3CA mutations (Sequist
201166), MET amplification (Engelman 200768)
Melanoma NF1 loss (Whittaker 201369 and Maertens 201370), BRAF amplification (Corcoran 201072), NRAS
PTEN loss (Paraiso 201171) amplification, increase in CRAF (Montagut
200873), splice variant BRAF (Poulikakos
201174), increased activation of AKT (Atefi
201175), NF1 loss (Whittaker 201369)
Colorectal EGFR activation (Prahallad 201276 and Corcoran NA
201277), PI3K/AKT activation (Mao 201378), Wnt/
Ca21 activation (Spreafico 201379)
Ovarian PI3K activation (Sheppard 201380), activation of NA
ERa (Hou 201381)

Abbreviations: BIM, B-cell chronic lymphocytic leukemia/lymphoma (Bcl-2)-like 11; BRAF, v-Raf murine sarcoma viral oncogene homolog B1; CRAF, RAF
proto-oncogene serine/threonine-protein kinase; EGFR, epidermal growth factor receptor; ERa, estrogen receptor a; Her2, human epidermal growth factor
receptor 2; MET, mesenchymal epithelial transition factor; NA, not applicable; NF1, nuclear factor 1; NRAS, neuroblastoma RAS viral oncogene homolog;
PI3K, phosphatidylinositol-3-kinase; PTEN, phosphatase and tensin homolog; T790M, threonine to methionine substitution at position 790; Wnt, wingless
type.

Figure 2. Mechanisms of resistance to v-raf murine sarcoma viral oncogene homolog B1 (B-Raf) inhibitors are illustrated. (a) In
melanoma, mechanisms of secondary resistance to BRAF inhibitors include the expression of BRAF splice variants, RAF proto-
oncogene serine/threonine-protein kinase (C-Raf) activation (all of which activates mitogen-activated protein kinase/extracellular
signal-regulating kinase [MAPK/ERK]), or signaling through alternative pathways like protein kinase B/mammalian target of rapa-
mycin (AKT/mTOR), etc (see Table 4). N-Ras indicates neuroblastoma RAS viral oncogene homolog; V600E, a valine to glutamic
acid substitution at BRAF position 600; PI3 kinase, phosphatidylinositol-3-kinase; MEK, mitogen-activated protein kinase-kinase.
(b) In colorectal cancer, primary resistance to BRAF inhibitors is caused by direct activation of epidermal growth factor receptor
(EGFR) and the AKT/mTOR pathway. CDC25C, indicates cell division cycle 25C; WNT, wingless type.

include low-grade serous and endometrioid, clear cell, had V600BRAF or KRAS codon 12 mutations. It is inter-
mucinous, and borderline tumors (BOTs); low-grade se- esting to note that, in 1 study, patients who had BRAF
rous tumors have mutations in KRAS (27%-36%), BRAF mutations had no recurrence after a median follow-up of
(33%-50%), and PIK3CA; whereas nearly all mucinous 3.6 years.98 Ho et al observed KRAS or BRAF mutations
tumors may have KRAS mutations.41,42,97 One study in 86% of cystadenomas adjacent to BOTs, and 88% of
reported that 57% of BOTs or low-grade serous tumors BOTs had mutations.99 There is a loss of frequency of

Cancer Month 00, 2014 7


Review Article

BRAF and KRAS mutations in the transition from nonma- signaling, is also regulated by modulation through parallel
lignant to malignant disease, from cystadenoma or BOT, pathways. This creates a complexity within and between
to invasive low-grade serous cancer. The mechanism of tumors that impedes the ability to translate therapeutic
this selective process, in which there is loss of an otherwise findings across tumors. Tissue and subtype specificity in
recognized oncogenic mutation during the process of ac- signaling adds a level of complexity to the application of
quisition of an invasive phenotype, is unknown. This is novel targeted agents, even against an otherwise dominant
the only example we identified in which there was such pathway. The MAPK/ERK pathway stimulates cellular
loss of a perceived gain-of-function mutation. proliferation and invasion; however, its activation also can
The identification of these mutations led to the logi- increase cellular apoptosis or antagonize pro-oncogenic
cal hypothesis that such ovarian neoplasms were a new input from other signals. The MAPK/ERK pathway dem-
frontier for experimentation with targeted BRAF and onstrates both oncogene and tumor suppressor, effects
MEK inhibitor therapy. Gynecologic Oncology Group depending on the tissue-specific tumor microenviron-
Study 239, which was a phase 2 trial of the MEK inhibitor ment. Although cancers share common mutations, differ-
selumetinib in 52 previously treated patients with ent cell types have developed unique responses to the
low-grade serous ovarian tumors, yielded a 15% overall mutations. These mutations may behave as oncogenic
response rate and a median progression-free survival of 11 drivers, passenger mutations, or regulatory events. The
months. This was compared with an historical role of the MAPK/ERK pathway in the tumor microen-
progression-free survival of 7 months. Mutational analy- vironment has long been recognized. This pathway is crit-
ses were performed in tumors from 34 patients, and KRAS ical in the process of physiologic and malignant invasion
and BRAF mutations were found in 41% and 6%, respec- and angiogenesis; and, most recently, a clear role for
tively, although mutations did not correlate with response MAPK/ERK has been demonstrated in tumor-immune
or longer progression-free survival.100 These examples system interactions. Hence, MAPK/ERK activation is a
argue against a preponderant role of the MAPK/ERK multifaceted target under varied regulatory bodies. Regu-
pathway as a targeting oncogenic driver in these tumors latory mechanisms may lead to the activation of alterna-
despite the presence of mutations. Studies of sorafenib tive pathways and paradoxical hyperactivation of the
predominantly in patients with recurrent, high-grade normal MAPK/ERK pathway. One unintended and
serous ovarian cancer did not demonstrate biochemical unexpected consequence of KRAS/BRAF inhibitor drug
activity of reduced ERK activation pretreatment or on- therapy is increased activity of the normal MAPK/ERK
treatment, perhaps consistent with the lack of genomic pathway, which can lead to the development of secondary
events in the MAPK/ERK pathway in those tumors.101 malignancies. Some novel combination therapies have
Interactions between the MAPK/ERK pathway and demonstrated increased treatment efficacy by addressing
estrogen receptor-a (ERa) have also been identified in both a specific target and its counter-regulatory effect in
preclinical studies. MEK inhibition caused an increase in the complex milieu of cellular signaling. In shaping future
ERa expression independent of AKT signaling in ovar- approaches toward personalized medicine, the challenge is
ian cell lines that were positive for ERa. The addition of clear: we must strike a delicate balance between exploiting
the ER inhibitor fulvestrant caused synergistic suppres- shared genetic targets and acknowledging the unique fea-
sion of tumor growth in vitro and in an in vivo model.81 tures of human cancers.
This may be a direction for clinical study using modula-
tion of the MAPK/ERK pathway to secondarily regulate FUNDING SUPPORT
a parallel pathway. This reinforces how ovarian cancer is This work was supported by the Intramural Program of the Center
a challenging environment in which to study the tumor- for Cancer Research, National Cancer Institute, National Institutes
of Health, USA.
specific effects of MAPK/ERK pathway activation. Its
broad range of cellular diversity and complexity of path-
CONFLICT OF INTEREST DISCLOSURES
way activation lends itself to combination therapy, neces-
The authors made no disclosures.
sitating greater understanding of the interaction of the
pathways. REFERENCES
1. Hudson TJ, Anderson W, Artez A, et al. International network of
Conclusion cancer genome projects. Nature. 2010;464:993-998.
2. Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N,
The downstream MAPK/ERK signaling node, which is Sander C. Emerging landscape of oncogenic signatures across
activated predominantly by upstream SRC/RAS/RAF human cancers. Nat Genet. 2013;45:1127-1133.

8 Cancer Month 00, 2014


Tissue-Specific MAPK Signaling/Burotto et al

3. Weinstein IB, Joe A. Oncogene addiction. Cancer Res. 2008;68: 28. Deschenes-Simard X, Gaumont-Leclerc MF, Bourdeau V, et al.
3077-3080; discussion 3080. Tumor suppressor activity of the ERK/MAPK pathway by promot-
4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next genera- ing selective protein degradation. Genes Dev. 2013;27:900-915.
tion. Cell. 2011;144:646-674. 29. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Onco-
5. De Luca A, Maiello MR, D’Alessio A, Pergameno M, Normanno genic ras provokes premature cell senescence associated with accu-
N. The RAS/RAF/MEK/ERK and the PI3K/AKT signalling path- mulation of p53 and p16INK4a. Cell. 1997;88:593-602.
ways: role in cancer pathogenesis and implications for therapeutic 30. Bric A, Miething C, Bialucha CU, et al. Functional identification
approaches. Expert Opin Ther Targets. 2012;16(suppl 2):S17-S27. of tumor-suppressor genes through an in vivo RNA interference
6. Yang SH, Sharrocks AD, Whitmarsh AJ. MAP kinase signalling screen in a mouse lymphoma model. Cancer Cell. 2009;16:324-
cascades and transcriptional regulation. Gene. 2013;513:1-13. 335.
7. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, 31. Colombino M, Capone M, Lissia A, et al. BRAF/NRAS mutation
Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546- frequencies among primary tumors and metastases in patients with
1558. melanoma. J Clin Oncol. 2012;30:2522-2529.
8. Tebbutt N, Pedersen MW, Johns TG. Targeting the ERBB family 32. Edlundh-Rose E, Egyhazi S, Omholt K, et al. NRAS and BRAF
in cancer: couples therapy. Nat Rev Cancer. 2013;13:663-673. mutations in melanoma tumours in relation to clinical characteris-
9. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat tics: a study based on mutation screening by pyrosequencing. Mela-
Rev Cancer. 2003;3:459-465. noma Res. 2006;16:471-478.
33. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene
10. Muram-Zborovski TM, Stevenson DA, Viskochil DH, Dries DC,
in human cancer. Nature. 2002;417:949-954.
Wilson AR, Rong M. SPRED 1 mutations in a neurofibromatosis
clinic. J Child Neurol. 2010;25:1203-1209. 34. Murugan AK, Dong J, Xie J, Xing M. MEK1 mutations, but not
ERK2 mutations, occur in melanomas and colon carcinomas, but
11. Wan PT, Garnett MJ, Roe SM, et al. Mechanism of activation of
none in thyroid carcinomas. Cell Cycle. 2009;8:2122-2124.
the RAF-ERK signaling pathway by oncogenic mutations of
B-RAF. Cell. 2004;116:855-867. 35. Nikolaev SI, Rimoldi D, Iseli C, et al. Exome sequencing identifies
recurrent somatic MAP2K1 and MAP2K2 mutations in melanoma.
12. Farooq A, Zhou MM. Structure and regulation of MAPK phospha-
Nat Genet. 2012;44:133-139.
tases. Cell Signal. 2004;16:769-779.
36. Seo JS, Ju YS, Lee WC, et al. The transcriptional landscape and muta-
13. Cossa G, Gatti L, Cassinelli G, Lanzi C, Zaffaroni N, Perego P. tional profile of lung adenocarcinoma. Genome Res. 2012;22:2109-
Modulation of sensitivity to antitumor agents by targeting the 2119.
MAPK survival pathway. Curr Pharm Des. 2013;19:883-894.
37. Cardarella S, Ogino A, Nishino M, et al. Clinical, pathologic, and
14. Dhanasekaran N, Premkumar Reddy E. Signaling by dual specific- biologic features associated with BRAF mutations in non-small cell
ity kinases. Oncogene. 1998;17:1447-1455. lung cancer. Clin Cancer Res. 2013;19:4532-4540.
15. Robinson MJ, Cobb MH. Mitogen-activated protein kinase path- 38. Cancer Genome Atlas Network. Comprehensive molecular character-
ways. Curr Opin Cell Biol. 1997;9:180-186. ization of human colon and rectal cancer. Nature. 2012;487:330-
16. Pullikuth AK, Catling AD. Scaffold mediated regulation of MAPK 337.
signaling and cytoskeletal dynamics: a perspective. Cell Signal. 39. Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colo-
2007;19:1621-1632. rectal cancer. N Engl J Med. 2009;361:98-99.
17. Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. 40. Sieben NL, Macropoulos P, Roemen GM, et al. In ovarian neo-
Phosphorylation and functional inactivation of TSC2 by Erk implica- plasms, BRAF, but not KRAS, mutations are restricted to low-
tions for tuberous sclerosis and cancer pathogenesis. Cell. 2005;121: grade serous tumours. J Pathol. 2004;202:336-340.
179-193. 41. Bell DA. Origins and molecular pathology of ovarian cancer. Mod
18. Chu B, Soncin F, Price BD, Stevenson MA, Calderwood SK. Pathol. 2005;18(suppl 2):S19-S32.
Sequential phosphorylation by mitogen-activated protein kinase and 42. Singer G, Oldt R 3rd, Cohen Y, et al. Mutations in BRAF and
glycogen synthase kinase 3 represses transcriptional activation by KRAS characterize the development of low-grade ovarian serous
heat shock factor-1. J Biol Chem. 1996;271:30847-30857. carcinoma. J Natl Cancer Inst. 2003;95:484-486.
19. Schmid RS, Pruitt WM, Maness PF. A MAP kinase-signaling path- 43. Bansal M, Gandhi M, Ferris RL, et al. Molecular and histopatho-
way mediates neurite outgrowth on L1 and requires Src-dependent logic characteristics of multifocal papillary thyroid carcinoma. Am J
endocytosis. J Neurosci. 2000;20:4177-4188. Surg Pathol. 2013;37:1586-1591.
20. Wolf I, Rubinfeld H, Yoon S, Marmor G, Hanoch T, Seger R. 44. Xing M, Alzahrani AS, Carson KA, et al. Association between
Involvement of the activation loop of ERK in the detachment from BRAF V600E mutation and mortality in patients with papillary
cytosolic anchoring. J Biol Chem. 2001;276:24490-24497. thyroid cancer. JAMA. 2013;309:1493-1501.
21. Zassadowski F, Rochette-Egly C, Chomienne C, Cassinat B. Regu- 45. Cohen Y, Xing M, Mambo E, et al. BRAF mutation in papillary
lation of the transcriptional activity of nuclear receptors by the thyroid carcinoma. J Natl Cancer Inst. 2003;95:625-647.
MEK/ERK1/2 pathway. Cell Signal. 2012;24:2369-2377. 46. Xi L, Arons E, Navarro W, et al. Both variant and IGHV4-34-
22. Sigoillot FD, Evans DR, Guy HI. Growth-dependent regulation of expressing hairy cell leukemia lack the BRAF V600E mutation.
mammalian pyrimidine biosynthesis by the protein kinase A and Blood. 2012;119:3330-3332.
MAPK signaling cascades. J Biol Chem. 2002;277:15745-15751. 47. Tiacci E, Trifonov V, Schiavoni G, et al. BRAF mutations in
23. Arthur JS, Ley SC. Mitogen-activated protein kinases in innate im- hairy-cell leukemia. N Engl J Med. 2011;364:2305-2315.
munity. Nat Rev Immunol. 2013;13:679-692. 48. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-acti-
24. Winters ME, Mehta AI, Petricoin EF 3rd, Kohn EC, Liotta LA. vated protein kinase cascade for the treatment of cancer. Oncogene.
Supra-additive growth inhibition by a celecoxib analogue and 2007;26:3291-3310.
carboxyamido-triazole is primarily mediated through apoptosis. 49. Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity
Cancer Res. 2005;65:3853-3860. in the clinic. Nature. 2013;501:355-364.
25. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J. Molecular 50. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF
interpretation of ERK signal duration by immediate early gene inhibitors transactivate RAF dimers and ERK signalling in cells
products. Nat Cell Biol. 2002;4:556-564. with wild-type BRAF. Nature. 2010;464:427-430.
26. Murphy T, Hori S, Sewell J, Gnanapragasam VJ. Expression and 51. Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutation predicts
functional role of negative signalling regulators in tumour develop- sensitivity to MEK inhibition. Nature. 2006;439:358-362.
ment and progression. Int J Cancer. 2010;127:2491-2499. 52. Bollag G, Hirth P, Tsai J, et al. Clinical efficacy of a RAF inhibitor
27. Katz M, Amit I, Yarden Y. Regulation of MAPKs by growth factors and needs broad target blockade in BRAF-mutant melanoma. Nature.
receptor tyrosine kinases. Biochim Biophys Acta. 2007;1773:1161-1176. 2010;467:596-599.

Cancer Month 00, 2014 9


Review Article

53. Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibi- 75. Atefi M, von Euw E, Attar N, et al. Reversing melanoma cross-
tion enhances T-cell recognition of melanoma without affecting resistance to BRAF and MEK inhibitors by co-targeting the AKT/
lymphocyte function. Cancer Res. 2010;70:5213-5219. mTOR pathway [serial online]. PLoS One. 2011;6:e28973.
54. Bekaii-Saab T, Phelps MA, Li X, et al. Multi-institutional phase II 76. Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon
study of selumetinib in patients with metastatic biliary cancers. J cancer to BRAF(V600E) inhibition through feedback activation of
Clin Oncol. 2011;29:2357-2363. EGFR. Nature. 2012;483:100-103.
55. Ho AL, Grewal RK, Leboeuf R, et al. Selumetinib-enhanced radioio- 77. Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-
dine uptake in advanced thyroid cancer. N Engl J Med. 2013;368:623- activation of MAPK signaling contributes to insensitivity of BRAF
632. mutant colorectal cancers to RAF inhibition with vemurafenib.
56. Janne PA, Shaw AT, Pereira JR, et al. Selumetinib plus docetaxel Cancer Discov. 2012;2:227-235.
for KRAS-mutant advanced non-small-cell lung cancer: a rando- 78. Mao M, Tian F, Mariadason JM, et al. Resistance to BRAF inhibi-
mised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. tion in BRAF-mutant colon cancer can be overcome with PI3K in-
2013;14:38-47. hibition or demethylating agents. Clin Cancer Res. 2013;19:657-
57. Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600- 667.
mutant advanced melanoma treated with vemurafenib. N Engl J 79. Spreafico A, Tentler JJ, Pitts TM, et al. Rational combination of a
Med. 2012;366:707-714. MEK inhibitor, selumetinib, and the Wnt/calcium pathway
modulator, cyclosporin A, in preclinical models of colorectal can-
58. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-
cer. Clin Cancer Res. 2013;19:4149-4162.
mutated metastatic melanoma: a multicentre, open-label, phase 3
80. Sheppard KE, Cullinane C, Hannan KM, et al. Synergistic inhibi-
randomised controlled trial. Lancet. 2012;380:358-365.
tion of ovarian cancer cell growth by combining selective PI3K/
59. Vin H, Ching G, Ojeda SS, et al. Sorafenib suppresses JNK- mTOR and RAS/ERK pathway inhibitors. Eur J Cancer. 2013;49:
dependent apoptosis through inhibition of ZAK. Mol Cancer Ther. 3936-3944.
2014;13:221-229. 81. Hou JY, Rodriguez-Gabin A, Samaweera L, et al. Exploiting MEK
60. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and inhibitor-mediated activation of ERalpha for therapeutic interven-
MEK inhibition in melanoma with BRAF V600 mutations. N Engl tion in ER-positive ovarian carcinoma [serial online]. PLoS One.
J Med. 2012;367:1694-1703. 2013;8:e54103.
61. US Food and Drug Administration. Trametinib and Dabrafenib 82. Thomas RK, Baker AC, Debiasi RM, et al. High-throughput
approval. Washington, DC: US Food and Drug Administration; oncogene mutation profiling in human cancer. Nat Genet. 2007;39:
2014. 347-351.
62. Turke AB, Zejnullahu K, Wu YL, et al. Preexistence and clonal 83. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epi-
selection of MET amplification in EGFR mutant NSCLC. Cancer dermal growth factor receptor underlying responsiveness of non-
Cell. 2010;17:77-88. small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129-
63. Ng KP, Hillmer AM, Chuah CT, et al. A common BIM deletion 2139.
polymorphism mediates intrinsic resistance and inferior responses 84. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung
to tyrosine kinase inhibitors in cancer. Nat Med. 2012;18:521-528. adenocarcinomas to gefitinib or erlotinib is associated with a second
64. Nakagawa T, Takeuchi S, Yamada T, et al. EGFR-TKI resistance mutation in the EGFR kinase domain [serial online]. PLoS Med.
due to BIM polymorphism can be circumvented in combination 2005;2:e73.
with HDAC inhibition. Cancer Res. 2013;73:2428-2434. 85. Yang JY, Hung MC. A new fork for clinical application: targeting
65. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and forkhead transcription factors in cancer. Clin Cancer Res. 2009;15:
resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 752-757.
2005;352:786-792. 86. Fedorenko IV, Paraiso KH, Smalley KS. Acquired and intrinsic
66. Sequist LV, Waltman BA, Dias-Santagata D, et al. Genotypic and BRAF inhibitor resistance in BRAF V600E mutant melanoma. Bio-
histological evolution of lung cancers acquiring resistance to EGFR chem Pharmacol. 2011;82:201-209.
inhibitors [serial online]. Sci Transl Med. 2011;3:75ra26. 87. Wang H, Daouti S, Li WH, et al. Identification of the
67. Takezawa K, Pirazzoli V, Arcila ME, et al. HER2 amplification: a MEK1(F129L) activating mutation as a potential mechanism of
potential mechanism of acquired resistance to EGFR inhibition in acquired resistance to MEK inhibition in human cancers carrying
EGFR-mutant lung cancers that lack the second-site EGFRT790M the B-RafV600E mutation. Cancer Res. 2011;71:5535-5545.
mutation. Cancer Discov. 2012;2:922-933. 88. Roskoski R Jr. RAF protein-serine/threonine kinases: structure and
regulation. Biochem Biophys Res Commun. 2010;399:313-317.
68. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplifica-
tion leads to gefitinib resistance in lung cancer by activating 89. Rosell R, Bivona TG, Karachaliou N. Genetics and biomarkers in
ERBB3 signaling. Science. 2007;316:1039-1043. personalisation of lung cancer treatment. Lancet. 2013;382:720-
731.
69. Whittaker SR, Theurillat JP, Van Allen E, et al. A genome-scale
90. Antony R, Emery CM, Sawyer AM, Garraway LA. C-RAF
RNA interference screen implicates NF1 loss in resistance to RAF
mutations confer resistance to RAF inhibitors. Cancer Res. 2013;73:
inhibition. Cancer Discov. 2013;3:350-362.
4840-4851.
70. Maertens O, Johnson B, Hollstein P, et al. Elucidating distinct 91. Romano E, Pradervand S, Paillusson A, et al. Identification of mul-
roles for NF1 in melanoma genesis. Cancer Discov. 2013;3:338- tiple mechanisms of resistance to vemurafenib in a patient with
349. BRAFV600E-mutated cutaneous melanoma successfully rechal-
71. Paraiso KH, Xiang Y, Rebecca VW, et al. PTEN loss confers lenged after progression. Clin Cancer Res. 2013;19:5749-5757.
BRAF inhibitor resistance to melanoma cells through the suppres- 92. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell
sion of BIM expression. Cancer Res. 2011;71:2750-2760. plasticity. Nature. 2013;501:328-337.
72. Corcoran RB, Dias-Santagata D, Bergethon K, Iafrate AJ, 93. Di Nicolantonio F, Martini M, Molinari F, et al. Wild-type BRAF
Settleman J, Engelman JA. BRAF gene amplification can promote is required for response to panitumumab or cetuximab in meta-
acquired resistance to MEK inhibitors in cancer cells harboring the static colorectal cancer. J Clin Oncol. 2008;26:5705-5712.
BRAF V600E mutation [serial online]. Sci Signal 2010;3:ra84. 94. Richman SD, Seymour MT, Chambers P, et al. KRAS and BRAF
73. Montagut C, Sharma SV, Shioda T, et al. Elevated CRAF as a mutations in advanced colorectal cancer are associated with poor
potential mechanism of acquired resistance to BRAF inhibition in prognosis but do not preclude benefit from oxaliplatin or irinotecan:
melanoma. Cancer Res. 2008;68:4853-4861. results from the MRC FOCUS trial. J Clin Oncol. 2009;27:5931-
74. Poulikakos PI, Persaud Y, Janakiraman M, et al. RAF inhibitor 5937.
resistance is mediated by dimerization of aberrantly spliced 95. Kohn EC, Romano S, Lee JM. Clinical implications of using molecular
BRAF(V600E). Nature. 2011;480:387-390. diagnostics for ovarian cancers. Ann Oncol. 2013;24(suppl 10):x22-x26.

10 Cancer Month 00, 2014


Tissue-Specific MAPK Signaling/Burotto et al

96. Lee JM, Ledermann JA, Kohn EC. PARP Inhibitors for BRCA1/2 99. Ho CL, Kurman RJ, Dehari R, Wang TL, Shih Ie M. Mutations
mutation-associated and BRCA-like malignancies. Ann Oncol. of BRAF and KRAS precede the development of ovarian serous
2014;25:32-40. borderline tumors. Cancer Res. 2004;64:6915-6918.
100. Farley J, Brady WE, Vathipadiekal V, et al. Selumetinib in women
97. Shih Ie M, Kurman RJ. Ovarian tumorigenesis: a proposed model
with recurrent low-grade serous carcinoma of the ovary or perito-
based on morphological and molecular genetic analysis. Am J
neum: an open-label, single-arm, phase 2 study. Lancet Oncol.
Pathol. 2004;164:1511-1518.
2013;14:134-140.
98. Grisham RN, Iyer G, Garg K, et al. BRAF mutation is associated 101. Matei D, Sill MW, Lankes HA, et al. Activity of sorafenib in
with early stage disease and improved outcome in patients with recurrent ovarian cancer and primary peritoneal carcinomatosis: a
low-grade serous ovarian cancer. Cancer. 2013;119:548-554. gynecologic oncology group trial. J Clin Oncol. 2011;29:69-75.

Cancer Month 00, 2014 11

You might also like