<<

Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ERBBal Remedies: Combination Therapy for EGFR-mutant Lung Cancers

Pang-Dian Fan¹ and Helena A. Yu²,³

¹Department of Pathology and Human Oncology and Pathogenesis Program ²Division of Solid Tumor Oncology, Department of Medicine Memorial Sloan Kettering Cancer Center, New York, NY 10065 ³Weill Cornell Medical College, New York, NY 10065

Pang-Dian Fan has no conflict of interest to report. Helena A. Yu has research funding to her institution from Pfizer Oncology, Daiichi Oncology, AstraZeneca, and Novartis.

Running Title: Combination ERBB Therapy for EGFR-mutant Lung Cancers

Corresponding author: Helena Yu 300 East 66th Street New York, NY, 10065 Phone: 646 888 4274 Email: [email protected]

Summary: Multiple members of the ERBB/HER family of receptor tyrosine kinases have been implicated in mediating acquired resistance to EGFR inhibitors that are used to treat EGFR-mutant lung cancers. New single-agent and combination therapies targeting ERBB/HER family members are being investigated to either prevent or overcome the emergence of acquired resistance.

1

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Main body text: In this issue of Clinical Cancer Research, Romaniello and colleagues (1) describe a combination of clinically approved therapies that target two members of the ERBB/HER family of receptor tyrosine kinases – the epidermal receptor (EGFR) and HER2 (ERBB2) - and can overcome resistance to EGFR inhibition in an animal model of EGFR-mutant lung cancers.

Initial treatment with an EGFR inhibitor is the standard of care for non-small cell lung cancers (NSCLC) harboring activating mutations in EGFR. Four small molecule inhibitors (TKIs) spanning three generations of drug development have been approved by the Food and Drug Administration (FDA) for first-line therapy of EGFR- mutant lung cancers. Despite high initial response rates to (first generation), (first), and (second), the development of acquired resistance typically ensues, and has emerged in response to treatment with (third) as well. The mechanisms underlying acquired resistance frequently result in reactivation of the kinase activity of EGFR or activation of bypass signaling tracts that promote tumor growth and survival (2). Reactivation of EGFR kinase activity occurs through secondary alterations in EGFR that alter drug binding, such as the T790M mutation that confers resistance to first- and second-generation EGFR TKIs and the C797S mutation that emerges upon treatment with third-generation EGFR inhibitors. Commonly activated bypass signaling tracts include HER2 and the MET, both most often driven mechanistically by amplification of their respective genes. In resistance that is associated with MET amplification, downstream activation of the PI3K pathway is critically dependent on HER3 (ERBB3), yet another member of the ERBB/HER family.

Given the prominent roles of EGFR, HER2, and HER3 signaling in promoting resistance to EGFR inhibitors, simultaneous targeting of all three of these ERBB/HER family members is an attractive therapeutic strategy to investigate. The authors previously reported that a combination of three monoclonal antibodies against EGFR, HER2, and HER3 suppressed compensatory feedback loops triggered by targeting EGFR alone, and markedly reduced the growth of xenografts of erlotinib-resistant PC9ER cells that harbor both an EGFR exon 19 deletion and EGFR T790M (3). They subsequently demonstrated that a similar triplet of monoclonal antibodies promoted degradation of all three receptors, induced cellular senescence, and overcame resistance to osimertinib in cells with an established EGFR C797S mutation (4). Relapsed outgrowth of PC9ER xenograft tumors was seen after discontinuation of osimertinib administered at doses high enough to eradicate measurable tumors but lower than levels corresponding to therapeutic doses in human patients, but no outgrowth was observed when the triplet of antibodies was added to the same subtherapeutic doses of osimertinib. The authors have now simplified the treatment regimen to include only FDA-approved therapies targeting EGFR and HER2, and demonstrate that continuous or intermittent therapy with a combination of subtherapeutic doses of osimertinib with the monoclonal antibodies (anti- EGFR) and (anti-HER2) durably prevented relapses in PC9ER xenografts (1). The addition of the two antibodies to low-dose osimertinib also effectively eliminated relapses that emerged after treatment with low-dose osimertinib alone.

2

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Combination therapy against EGFR and HER2 has been investigated before using afatinib (a potent tyrosine kinase inhibitor of both EGFR and HER2) together with cetuximab to treat acquired resistance to first-generation EGFR TKIs. Preclinical studies using transgenic mouse models and cell line xenografts showed dramatic shrinkage of tumors harboring both the activating EGFR L858R mutation and the T790M mutation with the combination of afatinib and cetuximab, not with either drug alone (5). In a phase 1b of afatinib and cetuximab that included 126 patients with EGFR-mutant lung cancers resistant to either erlotinib or gefitinib, the overall objective response rate was 29% and median progression-free survival was 4.7 months, and the objective response rates were comparable in T790M-positive and negative tumors (6).

Would the combination of low-dose osimertinib, cetuximab, and trastuzumab be expected to exhibit more robust clinical activity in this setting? The most obvious difference between the two regimens is the mode of inhibition of HER2. While afatinib inhibits the kinase activity of HER2, administration of trastuzumab leads to increased endocytosis and degradation of the receptor. As expected, treatment with cetuximab and trastuzumab together results in degradation of both EGFR and HER2 in pre-established PC9ER xenografts, but Romaniello and colleagues have also shown that this combination dramatically lowers the levels of HER3, MET, and AXL, another receptor tyrosine kinase implicated in acquired resistance to EGFR inhibition. The mechanism underlying the reduced levels of these receptors remains unclear, but the finding provides an intriguing preclinical rationale for why the combination of low-dose osimertinib, cetuximab, and trastuzimab might exhibit greater clinical activity than the pairing of afatinib and cetuximab in the treatment of acquired resistance to EGFR TKIs.

The striking response of PC9ER xenografts to the novel three-drug regimen both warrants and requires further validation. Although the authors have previously shown that the C797S mutation can be identified in PC9ER tumors that have relapsed on subtherapeutic osimertinib, a single xenograft model cannot faithfully replicate the full spectrum of mechanisms that have emerged in clinical cases of resistance to osimertinib. In the setting of osimertinib resistance, there have been reports of more off-target resistance mechanisms such as RET and ALK fusions (7) and BRAF mutations (8) that may be less amenable to multi-drug ERBB/HER family inhibition. Testing the three-drug combination in additional patient-derived and genetically engineered models will determine which molecular subsets of patients may respond to this treatment.

Other combinations of kinase inhibitors and antibodies that target the ERBB/HER family are actively being explored in the setting of acquired resistance to EGFR TKIs. In this context, it is important to note that the decision to utilize an anti-HER2 antibody instead of an anti-HER3 antibody in the three-drug regimen was based on the FDA-approved status of the trastuzumab, and not on any observed differences in preclinical activity. There are robust preclinical data highlighting the involvement of HER3 activation in resistance to EGFR inhibitors that have provided the basis for an ongoing clinical trial of a HER3 antibody drug conjugate, U3-1402, in patients with EGFR-mutant lung cancers with acquired resistance to EGFR TKIs (NCT03260491). In addition, Della Corte and colleagues recently demonstrated that osimertinib at therapeutic doses combined with

3

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cetuximab can reverse osimertinib resistance in multiple xenograft models (9). A clinical trial is currently investigating the use of osimertinib and another EGFR antibody, , in patients with EGFR-mutant lung cancers (NCT02496663)(Fig. 1).

In summary, the authors report efficacy of the novel combination of low-dose osimertinib, cetuximab and trastuzumab in in vitro and animal studies that suggests a role for the combination in the acquired resistance setting. The regimen appears to effectively block downstream EGFR signaling as well as multiple relevant bypass signaling pathways including MET, AXL and HER3. We look forward to seeing these works corroborated in additional preclinical studies. Ongoing studies assessing HER3 antibodies and osimertinib/EGFR antibody combinations in the acquired resistance setting may inform whether related combinations also have merit.

Figure 1. Tyrosine kinase inhibitors and antibodies or antibody drug conjugates in clinical use or under clinical investigation to target EGFR, HER2, and HER3 in EGFR-mutant lung cancers.

4

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Works Cited: 1. Romaniello D, Mazzeo L, Mancini M, Marrocco I, Noronha A, Kreitman MS, et al. A Combination Of Approved Antibodies Overcomes Resistance Of Lung Cancer To Osimertinib By Blocking Bypass Pathways. Clin Cancer Res 2018 doi 10.1158/1078-0432.CCR-18-0450. 2. Yu HA, Suzawa K, Jordan E, Zehir A, Ni A, Kim R, et al. Concurrent Alterations in EGFR-Mutant Lung Cancers Associated with Resistance to EGFR Kinase Inhibitors and Characterization of MTOR as a Mediator of Resistance. Clin Cancer Res 2018;24(13):3108-18 doi 10.1158/1078-0432.CCR-17-2961. 3. Mancini M, Gaborit N, Lindzen M, Salame TM, Dall'Ora M, Sevilla-Sharon M, et al. Combining three antibodies nullifies feedback-mediated resistance to erlotinib in lung cancer. Sci Signal 2015;8(379):ra53 doi 10.1126/scisignal.aaa0725. 4. Mancini M, Gal H, Gaborit N, Mazzeo L, Romaniello D, Salame TM, et al. An oligoclonal antibody durably overcomes resistance of lung cancer to third- generation EGFR inhibitors. EMBO Mol Med 2018;10(2):294-308 doi 10.15252/emmm.201708076. 5. Regales L, Gong Y, Shen R, de Stanchina E, Vivanco I, Goel A, et al. Dual targeting of EGFR can overcome a major drug resistance mutation in mouse models of EGFR mutant lung cancer. J Clin Invest 2009;119(10):3000-10 doi 10.1172/JCI38746. 6. Janjigian YY, Smit EF, Groen HJ, Horn L, Gettinger S, Camidge DR, et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov 2014;4(9):1036-45 doi 10.1158/2159-8290.CD-14-0326. 7. Offin M, Somwar R, Rekhtman N, Benayed R, Chang JC, Plodkowski A, et al. Acquired ALK and RET Gene Fusions as Mechanisms of Resistance to Osimertinib in EGFR Mutant Lung Cancers. JCO Precision Oncology 2018 (in press). 8. Ho CC, Liao WY, Lin CA, Shih JY, Yu CJ, Chih-Hsin Yang J. Acquired BRAF V600E Mutation as Resistant Mechanism after Treatment with Osimertinib. J Thorac Oncol 2017;12(3):567-72 doi 10.1016/j.jtho.2016.11.2231. 9. Della Corte CM, Ciaramella V, Cardone C, La Monica S, Alfieri R, Petronini PG, et al. Antitumor Efficacy of Dual Blockade of EGFR Signaling by Osimertinib in Combination With or Cetuximab in Activated EGFR Human NCLC Tumor Models. J Thorac Oncol 2018;13(6):810-20 doi 10.1016/j.jtho.2018.02.02

5

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1:

đƫ!014%) đƫ! %01)1) đƫġ ā đƫăġāąĀĂ Antibodies and antibody drug conjugates

EGFR HER2 HER3

đƫ.(+0%*% đƫ"0%*% Tyrosine đƫ!ü0%*% đƫ+6%+0%*% kinase đƫ"0%*% đƫăĂĈĉĉ inhibitors đƫ/%)!.0%*% đƫ +)%0%*% đƫ+6%+0%*% đƫ6.0%*% đƫăĂĈĉĉ

© 2018 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 22, 2018; DOI: 10.1158/1078-0432.CCR-18-2179 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ERBBal Remedies: Combination Therapy for EGFR-mutant Lung Cancers

Pang-Dian Fan and Helena A. Yu

Clin Cancer Res Published OnlineFirst August 22, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-18-2179

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2018/08/22/1078-0432.CCR-18-2179. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research.