Abstract

BackgroundThe benefit of precision medicine based on relatively limited gene sets and often-archived samples remains unproven. PERMED-01 (NCT02342158) was a prospective monocentric clinical trial assessing, in adults with advanced solid cancer, the feasibility and impact of extensive molecular profiling applied to newly biopsied tumor sample and based on targeted NGS (t-NGS) of the largest gene panel to date and whole-genome array-comparative genomic hybridization (aCGH) with assessment of single-gene alterations and clinically relevant genomic scores.MethodsEligible patients with refractory cancer had one tumor lesion accessible to biopsy. Extracted tumor DNA was profiled by t-NGS and aCGH. We assessed alterations of 802 “candidate cancer” genes and global genomic scores, such as homologous recombination deficiency (HRD) score and tumor mutational burden. The primary endpoint was the number of patients with actionable genetic alterations (AGAs). Secondary endpoints herein reported included a description of patients with AGA who received a “matched therapy” and their clinical outcome, and a comparison of AGA identification with t-NGS and aCGH versus whole-exome sequencing (WES).ResultsBetween November 2014 and September 2019, we enrolled 550 patients heavily pretreated. An exploitable complete molecular profile was obtained in 441/550 patients (80%). At least one AGA, defined in real time by our molecular tumor board, was found in 393/550 patients (71%, two-sided 90%CI 68–75%). Only 94/550 patients (17%, 95%CI 14–21) received an “AGA-matched therapy” on progression. The most frequent AGAs leading to “matched therapy” included PIK3CA mutations, KRAS mutations/amplifications, PTEN deletions/mutations, ERBB2 amplifications/mutations, and BRCA1/2 mutations. Such “matched therapy” improved by at least 1.3-fold the progression-free survival on matched therapy (PFS2) compared to PFS on prior therapy (PFS1) in 36% of cases, representing 6% of the enrolled patients. Within patients with AGA treated on progression, the use of “matched therapy” was the sole variable associated with an improved PFS2/PFS1 ratio. Objective responses were observed in 19% of patients treated with “matched therapy,” and 6-month overall survival (OS) was 62% (95%CI 52–73). In a subset of 112 metastatic breast cancers, WES did not provide benefit in term of AGA identification when compared with t-NGS/aCGH.ConclusionsExtensive molecular profiling of a newly biopsied tumor sample identified AGA in most of cases, leading to delivery of a “matched therapy” in 17% of screened patients, of which 36% derived clinical benefit. WES did not seem to improve these results.Trial registrationID-RCB identifier: 2014-A00966-41; ClinicalTrials.gov identifier: NCT02342158.

Highlights

  • The benefit of precision medicine based on relatively limited gene sets and often-archived samples remains unproven

  • Within patients with actionable genetic alterations (AGAs) treated on progression, the use of “matched therapy” was the sole variable associated with an improved PFS2/PFS1 ratio

  • In a subset of 112 metastatic breast cancers, whole-exome sequencing (WES) did not provide benefit in term of AGA identification when compared with t-next-generation sequencing (NGS)/ arraycomparative genomic hybridization (aCGH)

Read more

Summary

Introduction

The benefit of precision medicine based on relatively limited gene sets and often-archived samples remains unproven. The development of molecularly targeted therapies (MTT) directed against oncogenic drivers led to major progresses in the treatment of advanced cancers. More than 400 oncogenic drivers exist across cancers [1], but only a small fraction of them are targeted by the currently approved therapies; major research efforts are ongoing to develop MTT directed against the yet untargeted drivers. Most cancer types, including the most frequent, display a few drivers with relatively high frequency, and many drivers with very rare occurrence and shared with other cancer types. These potentially actionable very rare alterations provide opportunities for therapeutic targeting across different cancer types. Even if the functional value of an alteration depends on the cancer type [2], impressive tumor responses to therapies directed against a rare alteration have been reported in most cancers [2,3,4,5]

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.