TY - JOUR T1 - Genomic and Transcriptomic Determinants of Resistance to CDK4/6 Inhibitors and Response to Combined Exemestane plus Everolimus and Palbociclib in Patients with Metastatic Hormone Receptor Positive Breast Cancer JF - medRxiv DO - 10.1101/2022.07.11.22277416 SP - 2022.07.11.22277416 AU - Jorge Gómez Tejeda Zañudo AU - Romualdo Barroso-Sousa AU - Esha Jain AU - Qingchun Jin AU - Tianyu Li AU - Jorge E. Buendia-Buendia AU - Alyssa Pereslete AU - Daniel L. Abravanel AU - Arlindo R. Ferreira AU - Eileen Wrabel AU - Karla Helvie AU - Melissa E. Hughes AU - Ann H. Partridge AU - Beth Overmoyer AU - Nancy U. Lin AU - Nabihah Tayob AU - Sara M. Tolaney AU - Nikhil Wagle Y1 - 2022/01/01 UR - http://medrxiv.org/content/early/2022/07/12/2022.07.11.22277416.abstract N2 - Even though multiple resistance mechanisms and pathways for cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have been discovered, the complete landscape of resistance is still being elucidated. Moreover, the optimal subsequent therapy to overcome resistance remains uncertain.To address this, we carried out a phase I/II clinical trial of exemestane plus everolimus and palbociclib, triplet therapy for CDK4/6i-resistant hormone receptor–positive (HR+), HER2-metastatic breast cancer, one of the first trials evaluating CDK4/6i after CDK4/6i progression. With an observed clinical benefit rate of 18.8% (n = 6/32), the trial did not meet its primary efficacy endpoint. However, we leveraged the multi-omics tumor data from these patients to study the landscape of CDK4/6i resistance and to identify correlates of response to triplet therapy.We generated whole exome sequencing from 24 tumor and 17 ctDNA samples and transcriptome sequencing from 27 tumor samples obtained from 26 patients in the trial. Genomic and evolutionary analysis recapitulated the spectrum of known resistance genes (ERBB2, NF1, AKT1, RB1, ESR1) and pathways (RTK/MAPK, PI3K/AKT/mTOR, cell cycle, estrogen receptor), discovered potential new mechanisms of resistance in these pathways (ERBB2 amplification, BRAFV600E, MTORT1977R), and identified a patient with co-existing tumor lineages with distinct activating ERBB2 mutations, potentially the first case of convergent evolution of HER2 activation following CDK4/6i therapy. Joint genomic and transcriptomic analysis revealed that genomic resistance mechanisms were associated with transcriptomic features in their respective pathways, suggesting that transcriptomic features could be used to identify the pathways driving resistance. In particular, the mutually exclusive ESR1 and ERBB2/BRAF mutations, were each linked with high activity in distinct pathway signatures (estrogen receptor pathway vs RTK/MAPK pathway, respectively) and were exclusive to distinct molecular subtypes (Luminal A or Luminal B vs HER2-E, respectively). Overall, incorporating clinical and multi-omics features in CDK4/6i-resistant tumors enabled identification of known or putative drivers of resistance to the prior CDK4/6i and anti-estrogen therapies in nearly every patient (n = 22/23), including several patients in which transcriptomic features were the sole drivers. Genomic and transcriptomic features – particularly PI3K/AKT/mTOR mutations and/or high mTORC1 pathway activity - suggested that clinical benefit to combined estrogen receptor, CDK4/6, and mTOR inhibition was correlated with activation of the mTOR pathway.Our results illustrate how transcriptome sequencing provides complementary and additional information to genome sequencing, and how integrating both may help better identify patients likely to respond to CDK4/6i therapies.Significance Combined endocrine, CDK4/6 inhibitor, and mTOR inhibitor therapy showed limited benefit in patients with HR+ metastatic breast cancer who had progressed on a prior CDK4/6 inhibitor. Multi-omics analysis of tumors from this trial identified novel genomic and transcriptomic drivers of CDK4/6i resistance, known or putative drivers of resistance in 22/23 patients, and correlates of response to the trial therapy. Integrated genome and transcriptome sequencing may better identify factors that determine response to CDK4/6i therapy and help select optimal therapy.Competing Interest StatementR. Barroso-Sousa has received consulting fees from AstraZeneca, Eli Lilly, Libbs, Merck, Roche, and Zodiac; non-CME fees from Bard Access, BMS, Eli Lilly, Libbs, Merck, Novartis, Pfizer, and Roche; has carried out contracted research for Roche; and has received travel, accommodation, and expenses from Eli Lilly, Roche, Daichi Sankyo, and Merck. E. Jain is a current employee of Repare Therapeutics. J. E. Buendia-Buendia is a current employee of Cellarity. A.R. Ferreira has received honoraria from Bayer, Daiichi Sankyo, Novartis, and Roche; and has received travel grants from Roche. N.U. Lin has received consulting fees from Puma, Seattle Genetics, Daichii-Sankyo, AstraZeneca, Denali Therapeutics, Prelude Therapeutics, Olema Pharmaceuticals, Aleta BioPharma, Affinia Therapeutics, Voyager Therapeutics, and Janssen; has received institutional research support from Genentech, Pfizer, Merck, Seattle Genetics, Zion Pharmaceuticals, Olema Pharmaceuticals, and AstraZeneca; has stocks and other ownership interests in Artera Inc. N. Wagle has received consulting fees from Eli Lilly & Co, Relay Therapeutics; has carried out contracted research for Puma Biotechnology; has ownership interests in Relay Therapeutics. S.M. Tolaney has received consulting fees or has had an advisory role for Novartis, Pfizer, Merck, Eli Lilly, AstraZeneca, Genentech/Roche, Eisai, Sanofi, Bristol Myers Squibb, Seattle Genetics, Odonate Therapeutics, CytomX Therapeutics, Daiichi Sankyo, Athenex, Gilead, Mersana, Certara, Ellipses Pharma, 4D Pharma, OncoSec Medical Inc., BeyondSpring Pharmaceuticals, OncXerna, Zymeworks, Zentalis, Blueprint Medicines, Reveal Genomics, ARC Therapeutics, Infinity Therapeutics, Chugai Pharmaceuticals, Myovant, Zetagen, and Reveal Genomics; has received research funding from Genentech/Roche, Merck, Exelixis, Pfizer, Lilly, Novartis, Bristol Myers Squibb, Eisai, AstraZeneca, NanoString Technologies, Cyclacel, Nektar, Gilead, Sanofi, and Seattle Genetics. No disclosures were reported by the other authors.Clinical TrialNCT02871791Funding StatementThis work was supported by an ASPIRE grant from Pfizer (to S. Tolaney), Saverin Breast Cancer Research Fund (to Dana-Farber/Harvard Cancer Center), NCI Specialized Program of Research Excellence (SPORE) Grant 1P50CA168504 (to Dana-Farber/Harvard Cancer Center), Susan G. Komen CCR15333343 (to N. Wagle), The Breast Cancer Alliance (to N. Wagle), The Cancer Couch Foundation (to N. Wagle), and Twisted Pink (to N. Wagle).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Institutional review board (IRB) of Dana-Farber/Harvard Cancer Center (DF/HCC) gave ethical approval for this work.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesTumor and germline whole-exome sequencing data and RNA sequencing data generated and analyzed for this study are in the process of being deposited in dbGaP and will be available upon publication of the manuscript. Additional data generated in this study including clinical trial data, patient metadata, and tumor exome analysis are available in the Supplementary Data. ER -