Comprehensive Genomic Profiling

GCP can detect multiple biomarkers with a single assay

Detect Multiple Biomarkers in a Single Assay

CGP can detect biomarkers at nucleotide-level resolution and typically comprises all major genomic variant classes (single nucleotide variants, indels, copy number variants, fusions, and splice variants). Additionally, CGP can detect genomic signatures such as TMB and MSI (tumor mutational burden and microsatellite instability, respectively), maximizing the ability to find clinically actionable alterations.

consolidate biopsy sample testing

Consolidate Testing to Save Time and Precious Samples

CGP consolidates biomarker detection into a single multiplex assay, eliminating the need for sequential testing. With a single test, you can assess the most prevalent as well as rare biomarkers. By assessing all biomarkers at once, you may increase chances of finding an actionable alteration. This potentially provides faster results, limits the input of precious biopsy samples, and may reduce the risks and cost associated with rebiopsy.1,2

reveal more tumor composition insights with GCP

Identify Actionable Alterations

CGP can offer both actionable and potentially actionable results to help identify more effective therapeutic paths and innovative clinical trial options for cancer patients. When tissue biopsies are unavailable, CGP from liquid biopsy may provide helpful information about a tumor's genomic make-up. CGP using tissue and liquid biopsy together may reveal more insights into a tumor's composition.3,4

Percentage of Potentially Actionable Alterations

Multiple studies have demonstrated the ability of CGP to identify potentially clinically relevant genomic alterations, across different tumor types.

Potentially Actionable Variants Identified in Patient Samples Patient Cohort Author
93.5% Single center, prospective study with 339 patients. Refractory cancers, multiple types: ovarian (18%), breast (16%), sarcoma (13%), renal (7%), and others Wheler et al 20165
94.5% Prospective study with 100 patients, diverse-histology, rare, or poor-prognosis cancers Hirshfield et al 20166
36.7% Prospective study with 10,000 patients with advanced cancer aross a vast array of solid tumor types Zehir et al 20177
90% Retrospective study with 96 patients across multiple tumor types Reitsma et al 20198
71% 6832 NSCLC patients Suh et al 20169

The percent of actionable alterations identified in each study varies according to patient cohort, study type, CGP panel used, and criteria for categorizing a genomic alteration as actionable.

Data on file.

Marjolijn Ligtenberg

What Do the Experts Say About Comprehensive Genomic Profiling?

Listen to Marjolijn Ligtenberg, Pathologist, Laboratory of Tumor Genetics, Radboud University Medical Center, and others discuss the value CGP offers to patients and healthcare providers.

How CGP Compares to Other Sequencing Methods

CGP vs Single-Gene Assays

Single-gene assays are limited to a single biomarker. Many times these assays do not cover the entire gene sequence, with the risk of missing important gene alterations.13

An iterative single-gene testing approach can lead to tissue depletion and repeat biopsies.13,15,16

CGP vs Targeted Panels

Targeted panels typically offer coverage of specific genes instead of the entire coding sequence. As a result they can miss important alterations.7

A comprehensive single assay that assesses a wide range of biomarkers increases the chances of obtaining relevant information vs. targeted panels.

CGP vs Exome Sequencing

Not only can whole-exome sequencing be cost-prohibitive when developing personalized therapies, but it can also lead to inadequate coverage to detect important variants in lower frequencies due to the need for high amounts of sequencing.17-21

Ludovic Lacroix

There are a lot of very impressive novel therapies coming on the market with new fusions that we need to detect.

Ludovic Lacroix
Department of Medical Biology and Pathology
Institut de Cancerologie Gustave Roussy

At OmniSeq, we've adopted comprehensive genomic profiling as our test of choice so we could look at multiple biomarkers on a minimum of specimens to get a single consolidated report.

Jeff Conroy
Chief Scientific Officer at OmniSeq

A solid CGP assay is, in my opinion, inclusive of both DNA and RNA analysis. There is mounting data that clinically relevant variants can be missed by DNA analysis alone.

Nikoletta Sidiropoulos
Director of Molecular Pathology at the University of Vermont

The change from single-gene or single-biomarker testing to a comprehensive panel based approach has been driven by...factors that include inherent efficiency of a single comprehensive panel, which is key among cancers and other samples that have limited tissue.

Jeremy Wallentine
Laboratory Director at Intermountain Precision Genomics

It is important for CGP to include DNA and RNA targets. RNA fusions are very important in some cancers and you need to see the exact RNA fusion.

Professor Xiaoyan Zhou
Reference LabLeader of Molecular Pathological Lab Shanghai Cancer Center, Fudan University

Precision Medicine Through Comprehensive Genomic Profiling

NGS can power insights by examining hundreds of cancer genes in a single assay. Using CGP with liquid biopsies provides a quicker, less invasive, and less risky sample source for understanding complex cancers. Watch the video and learn how a complete suite of CGP offerings used within your lab can deliver faster precision medicine.

Watch the Video

Watch the video: Precision Medicine Through Comprehensive Genomic Profiling

Reasons to Bring CGP In-House

Adding comprehensive genomic profiling to your lab’s in-house test menu can provide a number of added benefits. These include:

icon fast results
Delivering results faster (as compared to send-out services)
icon reduce QNS rates
Reducing Quantity Not Sufficient (QNS) rates
icon increase informed cases
Increasing number of informed cases
icon database
Building a database for future studies
icon pathologist team
Amplifying the role of the pathologists on the care team
CGP Solution
CGP Solution

TruSight Oncology Comprehensive (EU) is the first CE-marked IVD test kit for comprehensive genomic profiling based on DNA and RNA that consolidates multiple iterative tests into one.

Learn More
Consult with an expert to learn more about CGP
Clinical Implications of Plasma-Based Genotyping With the Delivery of Personalized Therapy in Metastatic Non-Small Cell Lung Cancer

 

Genomic and Transcriptomic Profiling Expands Precision Cancer Medicine: the WINTHER Trial

 

Feasibility and Utility of a Panel Testing for 114 Cancer-associated Genes: A Hospital-based Study

 

References
  1. Pennel AP, Mutebi A, Zheng-Yi Z, et al. Economic Impact of Next-Generation Sequencing Versus Single-Gene Testing to Detect Genomic Alterations in Metastatic Non–Small-Cell Lung Cancer Using a Decision Analytic Model. JCO Precis Oncol. 2019. doi.org/10.1200/PO.18.00356.
  2. Lindeman NI, Cagle PT, Aisner DL, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn. 2018 Mar;20(2):129-159.
  3. Aggarwal C, Thompson JC, Black TA, et al.. Clinical Implications of Plasma-Based Genotyping With the Delivery of Personalized Therapy in Metastatic Non-Small Cell Lung Cancer. JAMA Oncol. 2019 Feb 1;5(2):173-180.
  4. Tukachinsky H, Madison RW, Chung JH, et al. Genomic analysis of circulating tumor DNA in 3,334 patients with advanced prostate cancer identifies targetable BRCA alterations and AR resistance mechanisms. Clin Cancer Res. 2021 Feb 8:clincanres.CCR-20-4805-E.2020.
  5. Wheler JJ, Janku F, Naing A et al Cancer Therapy Directed by Comprehensive Genomic Profiling: A Single Center Study. Cancer Res. 2016 Jul 1;76(13):3690-701.
  6. Hirshfield KM, Tolkunov D, Zhong H. Clinical Actionability of Comprehensive Genomic Profiling for Management of Rare or Refractory Cancers. Oncologist . 2016 Nov;21(11):1315-1325.
  7. Zehir A, Benayed R, Shah R et al Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med . 2017 Jun;23(6):703-713.
  8. Reitsma et al., 2019. Effect of a Collaboration Between a Health Plan, Oncology Practice, and Comprehensive Genomic Profiling Company from the Payer Perspective. Journal of Managed Care & Specialty Pharmacy. 2019 Jan 11:1-10
  9. Suh JH, Johnson A, Albacker L, et al. Comprehensive Genomic Profiling Facilitates Implementation of the National Comprehensive Cancer Network Guidelines for Lung Cancer Biomarker Testing and Identifies Patients Who May Benefit From Enrollment in Mechanism-Driven Clinical Trials. Oncologist. 2016 Jun;21(6):684-91.
  10. NIH National Cancer Institute. Cancer Drugs - National Cancer Institute. cancer.gov website. https://www.cancer.gov/about-cancer/treatment/types/targeted-therapies-fact-sheet. Accessed May 28, 2021.
  11. NIH US National Library of Medicine. Home - ClinicalTrials.gov. Find a study. https://www.clinicaltrials.gov/. Search terms included “genetic”, “genomic”, “DNA”, or “RNA”. Accessed January 25, 2021.
  12. Kopetz S, Shaw K, Lee J, et al. Use of a Targeted Exome Next-Generation Sequencing Panel Offers Therapeutic Opportunity and Clinical Benefit in a Subset of Patients With Advanced Cancers. JCO Precision Oncology. 2019;3:1-14.
  13. Drilon A, Wang L, Arcila ME, et al. Broad, Hybrid Capture-Based Next-Generation Sequencing Identifies Actionable Genomic Alterations in Lung Adenocarcinomas Otherwise Negative for Such Alterations by Other Genomic Testing Approaches. Clin Cancer Res. 2015;21(16):3631-363.
  14. Ali SM, Hensing T, Schrock AB, et al. Comprehensive Genomic Profiling Identifies a Subset of Crizotinib-Responsive ALK-Rearranged Non-Small Cell Lung Cancer Not Detected by Fluorescence In Situ Hybridization. Oncologist. 2016 Jun;21(6):762-70.
  15. Lim C, Tsao MS, Le LW, et al. Biomarker testing and time to treatment decision in patients with advanced nonsmall-cell lung cancer. Annals of Oncology. 2015;26(7):1415-1421.
  16. Yu TM, Morrison C, Gold EJ, et al. Multiple Biomarker Testing Tissue Consumption and Completion Rates With Single-gene Tests and Investigational Use of Oncomine Dx Target Test for Advanced Non-Small-cell Lung Cancer: A Single-center Analysis. Clin Lung Cancer. 2018 Jan;20(1):20-29.e8.
  17. Buchhalter I, Rempel E, Endris V, et al. Size matters: Dissecting key parameters for panel-based tumor mutational burden analysis. Int J Cancer. 2019;144(4):848-858 .
  18. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34.
  19. Pestinger V, Smith M, Sillo T, et al. Use of an integrated pan-cancer oncology enrichment NGS assay to Measure tumour mutational burden to detect clinically actionable variants. Mol Diagn Ther. 2020 Jun;24(3):339-349.
  20. Heydt C, Rehker J, Pappesch R, et al. Analysis of tumor mutational burden: correlation of five large gene panels with whole exome sequencing. Sci Rep. 2020 Jul 9;10(1):11387.
  21. Vanderwalde A, Spetzler D, Xiao N, et al. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med. 2018 Mar;7(3):746-756. Med. 2018 Mar;7(3):746-756.