erk inhibition: a new front in the war ... - cancer discovery · 140 | cancer discovery february...

4
VIEWS 140 | CANCER DISCOVERY FEBRUARY 2018 www.aacrjournals.org IN THE SPOTLIGHT ERK Inhibition: A New Front in the War against MAPK Pathway–Driven Cancers? Inna Smalley 1 and Keiran S.M. Smalley 1,2 1 Department of Tumor Biology, Moffitt Cancer Center & Research Insti- tute, Tampa, Florida. 2 Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, Florida. Corresponding Author: Keiran S.M. Smalley, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612. Phone: 813-745-8725; Fax: 813-449-8260; E-mail: keiran.smalley@moffitt.org doi: 10.1158/2159-8290.CD-17-1355 ©2018 American Association for Cancer Research. Summary: ERK inhibitors have enormous therapeutic potential against tumors that are BRAF mutant, BRAF–MEK inhibitor resistant, or RAS mutant. In this issue of Cancer Discovery, Sullivan and colleagues report on the first- in-human dose-escalation study of the ERK inhibitor ulixertinib, which they show to be well tolerated with clinical activity against a wide range of tumor types. Cancer Discov; 8(2); 140–2. ©2018 AACR. See related article by Sullivan et al., p. 184 (4). The MAPK pathway is the major signal transduction cas- cade that regulates cell growth; its activity is frequently upregulated in cancer. Under normal physiology, activation of the MAPK pathway occurs following the binding of growth factors to their cognate receptors, and the recruitment of the small GTPase RAS, which then recruits the serine threonine kinase RAF (1). This, in turn, activates MEK, which phos- phorylates ERK, the major effector kinase of the pathway. ERK then phosphorylates and activates multiple cytoplas- mic substrates and transcription factors (Fig. 1; ref. 1). The MAPK cascade is highly amplified at each signaling module, so that a relatively small input at the level of RAS leads to maximal stimulation of ERK. When activated in cancer, signaling through the MAPK pathway contributes to tumor progression through multiple mechanisms, including the maintenance of activated cyclin D1/CDK4 complexes (which increases cell growth), the suppression of proapoptotic mole- cules such as BIM (which increases cell survival), and through direct regulation of the cytoskeleton (which contributes to invasion and metastasis). The aberrant activation of the MAPK pathway observed in cancer occurs through multi- ple mechanisms, including amplification and overexpression of receptor tyrosine kinases (RTK), amplification of BRAF, mutations in RAS and BRAF, and the inactivation/mutations in tumor suppressors such as NF1. BRAF mutations occur in 7% of all cancers, and 50% of all cutaneous melanomas. To date, over 200 mutant BRAF alleles have been identified, and these have been subcatego- rized into three functional groups (2). The first two classes comprise the activating BRAF mutations that signal in a RAS-independent manner, as either active monomers (class I mutations, such as V600 mutations) or dimers (class II mutations, such as K601E/N/T, L597Q/V, and G469A/V/R; Fig. 1; ref. 2). The third class of BRAF mutations are kinase- dead and instead depend upon RAS for their signaling (class III, such as G466V/E, G469E, N581S, D594A/G/H; ref. 2). In cutaneous melanoma, the majority of BRAF mutations are V600E mutations, with a further 10% to 35% being non-V600 BRAF mutations. In other cancers, up to 50% of the BRAF mutations are non-V600. Among the three classes of BRAF mutations, only the class I are sensitive to BRAF inhibitors, with therapeutic strategies currently lacking for cancers with class II and class III BRAF mutations (2). Much of the MAPK- centric drug development to date has centered upon BRAF and MEK inhibitors, with the BRAF–MEK inhibitor combi- nation now FDA-approved for multiple BRAF V600 -dependent diseases, including melanoma, non–small cell lung cancer (NSCLC), and Erdheim–Chester disease. Although attempts were also made to develop MEK inhibitors for NRAS-mutant melanoma, this was halted on account of poor efficacy. Multiple clinical and preclinical studies have shown that BRAF inhibitors abrogate the growth and survival of mela- noma cells with BRAF V600E mutations. Although effective in the short term, therapeutic escape occurs, and this is often secondary to the relief of feedback inhibition in the MAPK pathway leading to increased RTK, RAS, and CRAF signal- ing and recovery of ERK signaling (Fig. 1). These effects can be overcome through vertical targeting of the MAPK pathway using the BRAF–MEK inhibitor combination, a strategy associated with an improved duration of antitumor response. In cases of acquired BRAF and BRAF–MEK inhibi- tor resistance, reactivation of the MAPK pathway is the most common mechanism of therapeutic escape, occurring in up to 70% of cases (3). It is now well established that melanoma cells have multiple mechanisms to reactivate the MAPK pathway and that even small changes in the MAPK signaling inputs can overcome the effects of BRAF inhibition. With this in mind, new strategies have been sought to suppress the recovery of MAPK signaling, with the goal of improving overall survival. In this issue of Cancer Discovery, Sullivan and colleagues report on the phase I clinical trial of the ERK inhibitor ulixertinib and provide the first clinical evidence that ERK inhibitors are active against cancers harboring diverse BRAF Research. on January 12, 2021. © 2018 American Association for Cancer cancerdiscovery.aacrjournals.org Downloaded from

Upload: others

Post on 21-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: eRK Inhibition: A new front in the War ... - Cancer Discovery · 140 | CANCER DISCOVERY February 2018 IN THE SPOTLIGHT eRK Inhibition: A new front in the War against MAPK Pathway–Driven

VIEWS

140 | CANCER DISCOVERY February 2018 www.aacrjournals.org

IN THE SPOTLIGHT

eRK Inhibition: A new front in the War against MAPK Pathway–Driven Cancers ? Inna Smalley 1 and Keiran S.M. Smalley 1 , 2

1 Department of Tumor Biology, Moffi tt Cancer Center & Research Insti-tute, Tampa, Florida. 2 Department of Cutaneous Oncology, Moffi tt Cancer Center & Research Institute, Tampa, Florida. Corresponding Author: Keiran S.M. Smalley , H. Lee Moffi tt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612. Phone: 813-745-8725; Fax: 813-449-8260; E-mail: keiran.smalley@moffi tt.org doi: 10.1158/2159-8290.CD-17-1355 ©2018 American Association for Cancer Research.

summary: ERK inhibitors have enormous therapeutic potential against tumors that are BRAF mutant, BRAF–MEK inhibitor resistant, or RAS mutant. In this issue of Cancer Discovery , Sullivan and colleagues report on the fi rst-in-human dose-escalation study of the ERK inhibitor ulixertinib, which they show to be well tolerated with clinical activity against a wide range of tumor types. Cancer Discov; 8(2); 140–2. ©2018 AACR.

See related article by Sullivan et al., p. 184 (4).

The MAPK pathway is the major signal transduction cas-cade that regulates cell growth; its activity is frequently upregulated in cancer. Under normal physiology, activation of the MAPK pathway occurs following the binding of growth factors to their cognate receptors, and the recruitment of the small GTPase RAS, which then recruits the serine threonine kinase RAF ( 1 ). This, in turn, activates MEK, which phos-phorylates ERK, the major effector kinase of the pathway. ERK then phosphorylates and activates multiple cytoplas-mic substrates and transcription factors ( Fig. 1 ; ref. 1 ). The MAPK cascade is highly amplifi ed at each signaling module, so that a relatively small input at the level of RAS leads to maximal stimulation of ERK. When activated in cancer, signaling through the MAPK pathway contributes to tumor progression through multiple mechanisms, including the maintenance of activated cyclin D1/CDK4 complexes (which increases cell growth), the suppression of proapoptotic mole-cules such as BIM (which increases cell survival), and through direct regulation of the cytoskeleton (which contributes to invasion and metastasis). The aberrant activation of the MAPK pathway observed in cancer occurs through multi-ple mechanisms, including amplifi cation and overexpression of receptor tyrosine kinases (RTK), amplifi cation of BRAF, mutations in RAS and BRAF, and the inactivation/mutations in tumor suppressors such as NF1.

BRAF mutations occur in 7% of all cancers, and 50% of all cutaneous melanomas. To date, over 200 mutant BRAF alleles have been identifi ed, and these have been subcatego-rized into three functional groups ( 2 ). The fi rst two classes comprise the activating BRAF mutations that signal in a RAS-independent manner, as either active monomers (class I mutations, such as V600 mutations) or dimers (class II

mutations, such as K601E/N/T, L597Q/V, and G469A/V/R; Fig. 1 ; ref. 2 ). The third class of BRAF mutations are kinase-dead and instead depend upon RAS for their signaling (class III, such as G466V/E, G469E, N581S, D594A/G/H; ref. 2 ). In cutaneous melanoma, the majority of BRAF mutations are V600E mutations, with a further 10% to 35% being non-V600 BRAF mutations. In other cancers, up to 50% of the BRAF mutations are non-V600. Among the three classes of BRAF mutations, only the class I are sensitive to BRAF inhibitors, with therapeutic strategies currently lacking for cancers with class II and class III BRAF mutations ( 2 ). Much of the MAPK-centric drug development to date has centered upon BRAF and MEK inhibitors, with the BRAF–MEK inhibitor combi-nation now FDA-approved for multiple BRAF V600 -dependent diseases, including melanoma, non–small cell lung cancer (NSCLC), and Erdheim–Chester disease. Although attempts were also made to develop MEK inhibitors for NRAS-mutant melanoma, this was halted on account of poor effi cacy.

Multiple clinical and preclinical studies have shown that BRAF inhibitors abrogate the growth and survival of mela-noma cells with BRAF V600E mutations. Although effective in the short term, therapeutic escape occurs, and this is often secondary to the relief of feedback inhibition in the MAPK pathway leading to increased RTK, RAS, and CRAF signal-ing and recovery of ERK signaling ( Fig. 1 ). These effects can be overcome through vertical targeting of the MAPK pathway using the BRAF–MEK inhibitor combination, a strategy associated with an improved duration of antitumor response. In cases of acquired BRAF and BRAF–MEK inhibi-tor resistance, reactivation of the MAPK pathway is the most common mechanism of therapeutic escape, occurring in up to 70% of cases ( 3 ). It is now well established that melanoma cells have multiple mechanisms to reactivate the MAPK pathway and that even small changes in the MAPK signaling inputs can overcome the effects of BRAF inhibition. With this in mind, new strategies have been sought to suppress the recovery of MAPK signaling, with the goal of improving overall survival.

In this issue of Cancer Discovery , Sullivan and colleagues report on the phase I clinical trial of the ERK inhibitor ulixertinib and provide the fi rst clinical evidence that ERK inhibitors are active against cancers harboring diverse BRAF

Research. on January 12, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 2: eRK Inhibition: A new front in the War ... - Cancer Discovery · 140 | CANCER DISCOVERY February 2018 IN THE SPOTLIGHT eRK Inhibition: A new front in the War against MAPK Pathway–Driven

views

February 2018 CANCER DISCOVERY | 141

Figure 1.  An overview of the MAPK signaling pathway with points of therapeutic intervention indicated. Activation of ERK can occur as the result of increased RTK signaling, RAS mutations, BRAF V600 and non-V600 mutation, and MEK mutations. ERK is the major effector of the pathway and leads to the phosphorylation of multiple cytoplasmic substrates. Once active, it translocates to the nucleus and activates a number of key transcription factors. To date, a number of drugs have been targeted against the MAPK pathway, including BRAF inhibitors (BRAFi), MEK inhibitors (MEKi), and now the ERK inhibitor (ERKi) ulixertinib. Inhibition of BRAF frequently leads to relief of feedback inhibition that decreases expression of negative regulators, such as a Sprouty, that facilitate recovery of MAPK pathway signaling.

Cell surface

Cytoplasm

Sprouty

Sprouty Cyclin D1

RTK

RAS BRAFiVemurafenibDabrafenibEncorafenib

MEKiTrametinibBinimetinibCobimetinib

ERKiUlixertinib

BRAFV600E

BRAFL597QP

PBIM

MCL

RSK

P

P

P

MEK

CytoplasmictargetsERK

PERK

c-JUN

Nucleus

ETSELKc-FOS

NRASKRASHRAS

CRAF

mutations, as well as melanomas with BRAF–MEK inhibitor resistance (4). The clinical trial consisted of an accelerated 3 + 3 rapid dose-escalation cohort and 6 expansion cohorts. A total of 135 patients were recruited, with 108 of these being enrolled into the expansion cohort. At the recommended phase II dose of ulixertinib (600 mg), ERK was completely inhibited in whole blood (4). Among those who responded in the dose-escalation phase, the best response was a partial response in 3 of 25 patients, all of who harbored a BRAF mutation. Of note, one of these responding patients had previously failed vemurafenib and dabrafenib and continued to respond to ulixertinib for >24 months. A further 6 patients had stable disease for >6 months as their best response, including one patient with a bronchoalveolar NSCLC. In the dose-expansion cohort, 11 of 81 (14%) patients experienced a partial response. Of those who responded, 3 of 18 had

NRAS-mutant melanoma, 3 of 12 had BRAF-mutant NSCLC (including BRAF V600E and L597Q mutations), and 4 of 21 had other BRAF-mutant cancers, including (L485W-mutant gallbladder cancer, V600E-mutant glioblastoma multiforme, G469A-mutant head and neck cancer, G469A-mutant small-bowel cancer; ref. 4). The clinical activity of ulixertinib was consistent with prior preclinical data in which good anti-proliferative activity was primarily seen in cell lines with MAPK pathway mutations (defined here as RAS- or RAF-mutant). The spectrum of genotypes showing sensitivity to ERK inhibition was broader than that of the BRAF inhibi-tors, with ulixertinib being effective in cancers with both non-V600 BRAF mutations (particularly class II BRAF muta-tions) and in BRAF-mutant cancers other than melanoma. In many instances, this was the first report that these genotypes were clinically actionable. Whether or not this represents a

Research. on January 12, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 3: eRK Inhibition: A new front in the War ... - Cancer Discovery · 140 | CANCER DISCOVERY February 2018 IN THE SPOTLIGHT eRK Inhibition: A new front in the War against MAPK Pathway–Driven

Views

142 | CANCER DISCOVERY February 2018 www.aacrjournals.org

broader range of actionable genotypes than has been reported for MEK inhibitors (which also have activity against L597S BRAF-mutant melanoma) remains to be determined (5).

Ulixertinib was also active in the cohort of patients with melanoma who had failed BRAF–MEK inhibitor therapy, with 3 of 19 showing a partial response (4). These observa-tions were supported by xenograft and cell-culture studies in which ulixertinib showed efficacy against BRAF-mutant A375 melanoma cells engineered to express mediators of BRAF inhibitor resistance, such as the MEKQ56P mutation, as well as cell lines with RAS mutations (6). It therefore seems likely that ERK inhibitors may be of use in patients who have failed BRAF and BRAF–MEK inhibitor therapy, particularly when the resistant tumors remain addicted to the MAPK pathway. The potential effects of ulixertinib against BRAF and BRAF–MEK inhibitor resistance mediated through alternate resist-ance mechanisms, such as activation of the PTEN–PI3K–AKT pathway, were not investigated.

It is possible that the true magnitude of ulixertinib’s effects against BRAF and BRAF–MEK inhibitor resistance was not seen in the current study. It is known that patients who have failed BRAF inhibitor therapy can be successfully rechallenged with the BRAF–MEK inhibitor combination and that up to 32% can experience a second durable response, provided that there is a sufficient washout period (7). Mecha-nistic studies have shown that certain types of BRAF inhibi-tor resistance, such as that mediated through increased RTK signaling, are dependent upon continuous drug selection pressure and that removal of drug restores sensitivity (8). Most of the BRAF inhibitor–resistant patients reported here by Sullivan and colleagues had relatively brief washout peri-ods (∼6 weeks), and this may have been too short for sensitiv-ity to be reestablished.

The evolving experience of targeted cancer therapy devel-opment suggests that resistance is inevitable when kinase inhibitors are used in a monotherapy setting, and it is likely that ulixertinib will ultimately be used as part of a drug combination. There is already preclinical evidence that the combination of ulixertinib and the BRAF inhibitor dabrafenib improves responses in BRAF-mutant melanoma xenograft models, with curative responses observed in multiple mice (6). Similarly, the combination of an ERK inhibitor with a MEK inhibitor leads to enhanced antitumor responses in cell culture models of NRAS-mutant melanoma (9). As reactiva-tion of the MAPK pathway is the most common resistance mechanism to BRAF and BRAF–MEK inhibitor therapy, there is clearly a rationale for using ulixertinib with the BRAF–MEK inhibitor combination. As increased toxicity is a concern when drugs are combined, an argument can be made for using these three drugs on an intermittent dosing schedule, in which the inhibitors are administered in sequence, with drug holidays. This approach has already been explored in preclinical models and appeared successful at delaying resistance in melano-

mas for which resistance was mediated by the expansion of preexisting cells with BRAF amplification (10). Whether this would work for all melanomas, or just a subset that utilize this particular resistance mechanism, remains to be determined. The continued development of ulixertinib as an ERK inhibi-tor, with a tolerable safety profile, provides another avenue to treat MAPK-driven cancers that were not previously deemed actionable. This, along with the expanded possibility of new MAPK pathway–targeted drug combinations and dose sched-ules, also offers hope for patients whose tumors are driven by BRAFV600, non-V600 BRAF, and NRAS mutations.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

AcknowledgmentsThis work was supported by R21 CA198550, R21 CA216756, and

P50CA168536-05 from the NIH and a Career Development Award from the Melanoma Research Foundation.

Published online February 5, 2018.

RefeRenCes 1. Wellbrock C, Karasarides M, Marais R. The RAF proteins take centre

stage. Nat Rev Mol Cell Biol 2004;5:875–85. 2. Yao Z, Yaeger R, Rodrik-Outmezguine VS, Tao A, Torres NM, Chang

MT, et al. Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS. Nature 2017;548:234–8.

3. Shi HB, Hugo W, Kong XJ, Hong A, Koya RC, Moriceau G, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov 2014;4:80–93.

4. Sullivan RJ, Infante JR, Janku F, Wong DJL, Sosman JA, Keedy V, et al. First-in-class ERK1/2 inhibitor ulixertinib (BVD-523) in patients with MAPK mutant advanced solid tumors: results of a phase I dose-esca-lation and expansion study. Cancer Discov 2018;8:184–95.

5. Dahlman KB, Xia JF, Hutchinson K, Ng C, Hucks D, Jia PL, et al. BRAF(L597) mutations in melanoma are associated with sensitivity to MEK inhibitors. Cancer Discov 2012;2:791–7.

6. Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, et al. Targeting the MAPK signaling pathway in cancer: promising preclinical activity with the novel selective ERK1/2 inhibitor BVD-523 (ulixertinib). Mol Cancer Ther 2017;16:2351–63.

7. Schreuer M, Jansen Y, Planken S, Chevolet I, Seremet T, Kruse V, 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 2017;18:464–72.

8. Sun C, Wang L, Huang S, Heynen GJ, Prahallad A, Robert C, et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 2014;508:118–22.

9. Rebecca VW, Alicea GM, Paraiso KH, Lawrence H, Gibney GT, Smal-ley KS. Vertical inhibition of the MAPK pathway enhances therapeu-tic responses in NRAS-mutant melanoma. Pigment Cell Melanoma Res 2014;27:1154–8.

10. Xue Y, Martelotto L, Baslan T, Vides A, Solomon M, Mai TT, et al. An approach to suppress the evolution of resistance in BRAF(V600E)-mutant cancer. Nat Med 2017;23:929–37.

Research. on January 12, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 4: eRK Inhibition: A new front in the War ... - Cancer Discovery · 140 | CANCER DISCOVERY February 2018 IN THE SPOTLIGHT eRK Inhibition: A new front in the War against MAPK Pathway–Driven

2018;8:140-142. Cancer Discov   Inna Smalley and Keiran S.M. Smalley  Driven Cancers?−

ERK Inhibition: A New Front in the War against MAPK Pathway

  Updated version

  http://cancerdiscovery.aacrjournals.org/content/8/2/140

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerdiscovery.aacrjournals.org/content/8/2/140.full#ref-list-1

This article cites 10 articles, 4 of which you can access for free at:

  Citing articles

  http://cancerdiscovery.aacrjournals.org/content/8/2/140.full#related-urls

This article has been cited by 4 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  SubscriptionsReprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerdiscovery.aacrjournals.org/content/8/2/140To request permission to re-use all or part of this article, use this link

Research. on January 12, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from