PT - JOURNAL ARTICLE AU - Jacob G. Scott AU - Geoff Sedor AU - Michael W. Kattan AU - Jeffrey Peacock AU - G. Daniel Grass AU - Eric A. Mellon AU - Ram Thapa AU - Michael Schell AU - Anthony Waller AU - Sean Poppen AU - George Andl AU - Steven A. Eschrich AU - Thomas J. Dilling AU - William S. Dalton AU - Louis B. Harrison AU - Tim Fox AU - Javier F. Torres-Roca TI - Optimizing Clinical Outcome and Toxicity in Lung Cancer Using a Genomic Marker of Radiosensitivity AID - 10.1101/2020.01.09.20017046 DP - 2020 Jan 01 TA - medRxiv PG - 2020.01.09.20017046 4099 - http://medrxiv.org/content/early/2020/07/04/2020.01.09.20017046.short 4100 - http://medrxiv.org/content/early/2020/07/04/2020.01.09.20017046.full AB - Cancer sequencing efforts have demonstrated that cancer is the most complex and heterogeneous disease that affects humans. However, radiation therapy, one of the most common cancer treatments, is prescribed based on an empiric one-size-fits all approach. We propose that the field of radiation oncology is operating under an outdated null hypothesis: that all patients are biologically similar and should uniformly respond to the same dose of radiation. We have previously developed the Genomic Adjusted Radiation Dose (GARD), a method which accounts for biological heterogeneity and can be utilized to predict optimal RT dose for an individual patient. In this article, we utilize GARD to characterize the biological imprecision of one-size-fits-all RT dosing schemes which result in both over- and under-dosing for the majority of patients treated with RT. To elucidate this inefficiency, and therefore the opportunity for improvement using a personalized dosing scheme, we develop a patient-specific competing hazards-style mathematical model combining the canonical equations for tumor control (TCP) and normal tissue complication probabilities (NTCP). This model simultaneously optimizes tumor control and toxicity by personalizing RT dose using patient-specific genomics. Using data from two prospectively collected cohorts of patients with non-small-cell lung cancer, we validate the competing hazards model by demonstrating that it predicts the results of RTOG 0617. We show how 0617 failure can be explained by the biological imprecision of empiric uniform dose escalation which results in 80% of patients being over-exposed to normal tissue toxicity without potential tumor control benefit. In conclusion, our data reveals a tapestry of radiosensitivity heterogeneity, provides a biological framework that explains the failure of empiric RT dose escalation, and quantifies the opportunity to improve clinical outcomes in lung cancer by incorporating genomics into RT.Competing Interest StatementJGS and JTR are co-inventors of and own IP for GARD and RxRSI. JTR and SE are co-inventors of and IP for RSI. JTR and SE have stock in Cvergenx. JGS has stock options in Cvergenx. TF, GA and AW have shared interest and stock in Varian Medical Systems.Funding StatementNo external funding was received.Author DeclarationsAll relevant ethical guidelines have been followed; any necessary IRB and/or ethics committee approvals have been obtained and details of the IRB/oversight body are included in the manuscript.YesAll necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.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.YesPlease see attached spreadsheet.