OVERVIEW
Liquid biopsy has been used extensively in solid malignancies to detect actionable driver mutations, to monitor treatment response, to detect recurrence, to identify resistance mechanisms, and to prognosticate outcome. Although many liquid biopsy sequencing platforms are being used, only five test kits have received government approval. We review representative literature on these government-approved liquid biopsy kits, which are primarily used to detect EGFR mutation in lung cancer and RAS (KRAS, NRAS, BRAF) mutations in colorectal carcinoma. Another emerging use of single-gene liquid biopsy is to detect PIK3CA mutations and to understand resistance to hormonal blockade in breast and prostate cancers. The two most commonly used next-generation sequencing (NGS) liquid biopsy tests (Guardant 360, Guardant Health; Foundation-ACT, Foundation Medicine Inc.) are discussed. The ability and the applicability of NGS platform to detect tumor mutation burden are also addressed. Finally, the use of circulating tumor DNA (ctDNA) to detect minimal residual disease may be the most important use of ctDNA in the setting of tumor heterogeneity. The ability to identify “shedders” and “nonshedders” of ctDNA may provide important insight into the clinicopathologic characteristics of the tumor and portend important prognostic significance regarding survival.
The use of liquid biopsy (primarily from blood) in solid malignancy provides a convenient and safe way to detect the presence of actionable driver mutations, to assess the resistance mechanisms to actionable driver mutations, to monitor treatment response, to detect early recurrence, to serve as an adjuvant to radiologic imaging as post-treatment surveillance, and to prognosticate the outcome of cancer treatment. Cell-free DNA, including ctDNA, circulating tumor cells, and exosomes containing tumor microRNAs can all be detected by liquid biopsy. The biologic nature of ctDNA, the various sequencing platforms used in liquid biopsy, and the various utilities of liquid biopsy have recently been expertly and comprehensively reviewed by Wan and colleagues.1 The many sequencing platforms used in liquid biopsy can be broadly summarized as nondigital, digital, and NGS. The performances of these individual platform have been reviewed extensively.2–10 However, only five liquid biopsy test kits are approved by government agencies.
DETECTION OF SPECIFIC ACTIONABLE GENOMIC ALTERATIONS BY LIQUID BIOPSY
Detection of Activating EGFR Mutations in Lung Cancer
Cobas EGFR mutations test version 2 (EGFR del 19, L858R, T790M).
The Cobas test (Roche Molecular Diagnostics, Pleasonton, CA) is the only U.S. Food and Drug Administration (FDA)–approved liquid biopsy to detect the two most common activating epidermal growth factor receptor (EGFR) mutations (EGFR del19 and EGFR L858R) for the selection of EGFR tyrosine kinase inhibitor. It was subsequently approved for the detection of the most common acquired resistance mutation, EGFR T790M, after progression with first- or second-generation EGFR tyrosine kinase inhibitors for selection of osimertinib to treat patients with EGFR T790M–positive non–small cell lung cancer (NSCLC; Table 1).11–14 ENSURE, a randomized phase III trial comparing erlotinib to platinum/gemcitabine chemotherapy as first-line treatment of EGFR-positive NSCLC provided the basis for the approval of detection of EGFR del19 and EGFR L858R.15 Additional large-scale real-life prospective trials (ASSESS, Europe and Japan; and IGNITE, Russia and China/South Korea/Taiwan) studying the feasibility and testing the concordance of using Cobas liquid biopsy versus tumor have been completed.16,17 In the IGNITE study, the concordance between 2,561 matched tissue/cytology and plasma samples was 80.5%, sensitivity was 46.9%, and specificity was 95.6%.17 In the ASSESS study, the concordance of mutation status in 1,162 matched samples was 89%, sensitivity was 46%, specificity was 97%, positive predictive value was 78%, and negative predictive value was 90%. Two combined single-arm phase II studies of osimertinib provided the basis for the approval for the detection of EGFR T790M.18 A European study (APPLE) investigating the use of liquid biopsy to detect EGFR T790M mutation is ongoing.19
TABLE 1.
Approved Liquid Biopsy Test Kits for Detection of Activating and Resistance EGFR Mutations in Non–Small Cell Lung Cancer
Therascreen EGFR Plasma RGQ PCR Kit | Cobas EGFR Mutation Test v2 | AmoyDX Super-ARMS EGFR Mutation Detection Kit | |
---|---|---|---|
PCR Kit | |||
Approving Agency/Region | European Union 98/79, | U.S. FDA | Chinese FDA |
Conformitée Européene-ln Vitro Diagnostics directive | |||
Date of Approval | January 12, 2015 | June 1, 2016 (EGFR del 19, EGFR L858R) September 28, 2016 (EGFR T790M) | January 22, 2018 (EGFR del 19, EGFR L858R, EGFR T790M) |
Manufacturer | Qiagen | Roche | AmoyDx |
Sequencing Platform | Scorpion Amplification Refractory Mutation System | Scorpion Amplification Refractory Mutation System | Scorpion Amplification Refractory Mutation system |
Detectable Technology* | Analog (real-time PCR) | Analog (real-time PCR) | Analog (real-time PCR) |
MAF Quantification | Semiquantitative | Semiquantitative | Semiquantitative |
No. of EGFR Mutations Detected | 29 | 42 | 41 |
Major EGFR Mutations Detected | EGFR del 19 (19 different mutations) | EGFR del 19 (29 different mutations) | EGFR del 19 (29 different mutations) |
EGFR L858R | EGFR L858R (2 different mutations) | EGFR L858R | |
EGFR T790M | EGFR T790M | EGFR T790M | |
EGFR G719X (3 different mutations) | EGFR G719X (3 different mutations) | EGFR G719X (3 different mutations) | |
EGFR S761I | EGFR S761I | EGFR S761I | |
EGFR L861Q | EGFR L861Q | EGFR L861Q | |
EGFR exon 20 insertions (3 insertions) | EGFR exon 20 insertions (5 insertions) | EGFR exon 20 insertions (5 insertions) | |
Indication | EGFR del 19, | EGFR del 19 | EGFR del 19 |
EGFR L858R | EGFR L858R | EGFR L858R | |
EGFR Del 19 and EGFR L858R | EGFR T790M | EGFR T790M | |
Study(ies) Supporting Approval | IFUM | ENSURE (Y025121) | Single-center, single-arm study (First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China) |
Comparator Tissue Test | Therascreen EGFR test (tissue use) | Cobas EGFR mutation test v2 (tissue test) NGS on an lllumina MiSeq | AmoyDx EGFR 29 mutation detection kit (tissue) |
No. of Patients | 859 with tumors successfully screened | 601 with tumors successfully screened | 109 screened |
652 with successfully paired tumor/plasma analyzed | 431 with successfully paired tumor/plasma analyzed and validated | 61 with tissue positive for all EGFR mutations | |
217 enrolled | 50 with plasma positive for all EGFR mutations | ||
Plasma RGQ PCR Kit | Mutation on Test v2 | Detection Kit | |
Primary Data | Screening for all patients: | Screening for all patients | Of 61 patients positive for EGFR mutations from tumor, 50 were also detected by plasma |
Of 105 patients positive for EGFR mutations from tumor, 69 (65.7%) were detected by plasma | Of 210 patients positive for EGFR mutations from tumor, 161 (76.7%) were detected by plasma | Of 48 patients negative for EGFR mutations from tumor, 48 were also negative for EGFR mutations by plasma | |
Of 547 patients negative for EGFR mutations from tumor, 546 (99.8%) were negative for EGFR mutation by plasma | Of 221 patients negative for EGFR mutations from tumor, 217 (98.2%) were negative for EGFR mutation by plasma | ||
Screening for EGFR del 19 patients | |||
Of 120 patients positive for EGFR del 19 mutation from tumor, 97 (80.8%) were detected by plasma | |||
Of 311 patients negative for EGFR del 19 mutation from tumor, 307 (98.7%) were negative for EGFR mutation by plasma | |||
Screening for EGFR L858R mutation | |||
Sensitivity: Of 90 patients positive for EGFR L858R from tumor, 61 (67.8%) were detected by plasma | |||
Specificity: Of 341 patients negative for EGFR L858R from tumor, 338 (99.1%) were negative for EGFR L858R by plasma | |||
Performance Characteristics** | Sensitivity: 65.7% (95% CI, 55.8%–74.7%) Specificity: 99.8% (95% CI, 999%–100%) Concordance: 94.3% (95% CI, 92.3%–96.0%) PPV: 98.6% (95% CI, 92.3%–100%) NPV: 93.8% (95% CI, 91.5%–95.6%) |
Prevalence: 15% | Sensitivity: 82% (95% CI, 72.3%–91.6%) Specificity: 100% PPV: 81.4% (95% CI, 71.4%–91.3%) NPV: 100% |
PPV: 88.6% (95% CI, 79.7%–96.6%) | |||
NPV: 96.0% (95% CI, 94.39%–97.6%) | |||
Prevalence: 40% | |||
PPV: 96.8% (95% CI, 93.0%–99.4%) | Concordance: 89.9% (95% CI, 84.3%–95.6%) | ||
NPV: 86.4% (95% CI, 83.39%–89.4%) | |||
Prevalence: 50% | |||
PPV: 97.8% (95% CI, 95.0%–100%) | |||
NPV: 80.8% (95% CI, 77.49%–84.8%) | |||
Clinical Outcome Data | ORR (EGFR del 19 + L858R) | PFS benefit of erlotinib over chemotherapy (ITT population, n = 217) | ORR |
Tumor+/plasma+: 72.5% (95% CI, 61.0%–81.6%); tumor+/plasma−: 63.6% (95% CI, 46.6%–77.8%) | HR, 0.33 (95% CI, 0.23–0.47); log-rank test p < .0001 | All patients: 64.3% (95% CI, 48%–78%; n = 42) | |
ORR [EGFR del 19) | PFS benefit of erlotinib over chemotherapy (tumor+/plasma+; n = 137) | Tumor+/plasma+: 65.7% (95% CI, 48%–81%; n = 35) | |
Tumor+/plasma+: 82.2% (95% CI, 68.7%–90.7%); tumor+/plasma−: 65.0% (95% CI, 43.5%–73.7%) | HR, 0.29 (95% CI, 0.19–0.45); log-rank test p < .0001 | Tumor+/plasma−: 57.15% (95% CI, 18%–90%) | |
ORR [EGFR L858R) | PFS benefit of erlotinib over chemo (tumor+/plasma−; n = 42) | ||
Median PFS (all enrolled patients) | HR, 0.37 (95% CI, 0.50–0.90) | ||
All enrolled patients: 9.7 mo (95% CI, 8.5–11.0 mo) | |||
Tumor+/plasma+: 10.2 mo (95% CI, 8.5–12.5 mo) | |||
EGFR del 19 | |||
All: 9.6 mo (95% CI, 8.0–11.0 mo) | |||
Tumor+/plasma+: 10.3 mo (95% CI, 8.5–12.4 mo) | |||
T790M | |||
Validation Study | NA | Combined AURA extension study and AURA2 (EGFR T790M) | AURA17 |
Primary Data | NA | 551 of 710 (78%) patients enrolled in both trials had paired tissue and plasma samples | 240 patients |
416 of 551 (75.5%) patients were plasma positive for T790M, 127 (23.0%) were plasma negative for T790M, and 8 patients had invalid plasma tests | |||
Comparator Tissue Test | Cobas EGFR mutation test v2 (tissue test) | Cobas EGFR mutation test v2 (tissue test) | |
Performance Parameters | NA | EGFR T790M | AmoyDx Super-ARMS EGFR T790M mutation detection kit only |
Sensitivity: 61% (95% CI, 57%–66%) | |||
Specificity: 79% (95% CI, 70%–85%) | EGFR T790M | ||
Concordance: 65% (95%: 61%–69%) | Sensitivity: 49% (95% CI, 41%–57%) | ||
EGFR del 19 | Specificity: 78% (95% CI, 67%–86%) | ||
Sensitivity: 85% (95% CI, 81%–89%) | |||
Specificity: 98% (95% CI, 95%–100%) | |||
Concordance: 90% (95% CI, 87%–92%) | |||
EGFR L858R | |||
Sensitivity: 76% (95% CI, 69%–82%) | |||
Specificity: 98% (95% CI, 96%–99%) | |||
Concordance: 91% (95% CI, 88%–93%) | |||
Clinical Outcome | NA | ORR | |
T790M tumor+/plasma+: 64% (95% CI, 57%–70%; n = 235) | |||
T790M tumor+/plasma−: 70% (95% CI, 62%–77%; n = 155) | |||
References | 20–25 | 11–13,18,23 | 26–28 |
Analog detection (also known as real-time PCR) uses fluorescent markers that attach to specific mutation sites, making them detectable; fluorescence in the sample is detected as a whole. In digital PCR, the sample is separated into many partitions, each tested individually, providing a lower threshold of detection.
Can detect the level of ctDNA at or above threshold limit, fully quantitative can measure the level ctDNA in a sample.
Abbreviations: FDA, U.S. Food and Drug Administration; PCR, polymerase chain reaction; MAF, mutant allele frequency; IFUM, Iressa Follow up Measure Study; NGS, next-generation sequencing; ORR, overall response rate; NPV, negative predictive value; PPV, positive predictive value; ITT, intention-to-treat; HR, hazard ratio; PFS, progression-free survival; NA, not applicable.
Therascreen EGFR plasma RGQ PCR kit.
The Therascreen EGFR Plasma RGQ PCR test (Qiagen Inc., Venlo, the Netherlands) is approved in the European Union (E.U.; Conformité Européene-In Vitro Diagnostics) for detection of the two most common EGFR mutations (EGFR del19 and EGFR L858R) but not T790M (Table 1).20–23 The IFUM study provided the basis for adoption of this test in European Union.24,25
AmoyDx Super-ARMS EGFR mutation test kit.
The newest government-approved liquid biopsy, the AmoyDx Super-ARMS EGFR mutation test kit (AmoyDx, Xiamen, China), was approved on January 22, 2018, by the Chinese Food and Drug Administration to detect EGFR del19, EGFR L858R, and EGFR T790M mutations (Table 1).26–28 AURA17 is a single-arm study of osimertinib in China that provided the supporting evidence for the approval of this test kit.28
Role in diagnosis.
Although all three tests can detect the three “uncommon” EGFR mutations (L861Q, G719X, and S768I)—for which afatinib was approved for use in the United States on January 16, 2018—none of them is officially approved to detect them.29 Table 1 shows that across all three test kits, the sensitivity for detecting T790M seems to be lower than that for detecting the two common activation mutations. The higher the prevalence of EGFR mutations (e.g., approximately 40%–50% in Asian populations), the lower the negative predictive value of the liquid biopsy (Table 1); this is based on the Cobas EGFR version 2 calculations. Thus, a negative EGFR plasma test result in a high-endemic region for EGFR mutations should be followed by a tumor tissue biopsy.
In addition, the National Institute of Health and Care Excellence in the United Kingdom recently published their technology summary of evidence of seven different liquid biopsy platforms.30 The four additional tests reviewed, in addition to the three government-approved tests, were the AmoyDx EGFR 29 mutation detection kit, AmoyDx Super-ARMS EGFR T790M mutation detection kit, Droplet Digital PCR (ddPCR) Dx system (Bio-Rad, Hercules, CA), and PANA Mutyper R EGFR kit (Panagene, Daejeon, Korea). The major summary finding is that plasma EGFR detection has specificity similar to but sensitivity lower than those of tumor biopsy. The resource impact to the health care system is similar to that for standard of care, but plasma testing could be cost-effective if this led to fewer tissue biopsies30
Going forward, increasing evidence suggests that the ability to identify detectable circulating EGFR mutations from liquid biopsy (“shedders” compared with “nonshedders”) has prognostic implications. In the FLAURA and AURA3 phase III trials (Table 2), although both shedders and nonshedders benefited from osimertinib, progression-free survival (PFS) in the osimertinib arm was numerically better for nonshedders than for shedders; this finding indicates that shedders in general have a higher tumor burden (Table 2).14,31 Additionally, among patients treated at National Taiwan University Hospital who progressed while receiving osimertinib, a retrospective genomic analysis that used plasma BEAMing technology (Sysmex Inostics, Hamburg, Germany) to detect EGFR mutations showed that, again, shedders had significantly shorter median PFS while receiving osimertinib, shorter PFS after progression on osimertinib, and shorter overall survival (OS) after osimertinib treatment (Table 2).32 Furthermore, 42.5% (17/40) of the shedders compared with only 7.7% (1/13) of nonshedders had brain metastases in this study, again indicating that shedders had more extensive/poor-prognosis disease than nonshedders. Finally, the “disappearance” of EGFR T790M at the time of progression during osimertinib treatment was associated with significantly shorter PFS after osimertinib therapy (Table 2), indicating more recalcitrant non–EGFR-mediated resistance mechanisms.32 Shedders of EGFR T790M usually have higher tumor burden with late-stage disease. Similarly, shorter PFS has been reported in shedders with EGFR L858R mutation than in nonshedders from a retrospective analysis of the EURTAC trial.33,34
TABLE 2.
Survival Outcome (Progression-Free Survival, Overall Survival, and Survival After Osimertinib Progression) of EGFR T790M Shedders Versus Nonshedders
FLAURA31 | |||||||||
---|---|---|---|---|---|---|---|---|---|
Overall (n = 556) | Shedders (n = 359) | Nonshedders (n = 124) | |||||||
Osiemrtinib | Gefitinib/Erlotinib | HR (95% CI); p Value | Osimertinib | Gefitinib/Erlotinib | HR (95%CI); p Value | Osimertinib | Gefitinib/Erlotinib | HR (95% CI); p Value | |
Patients, n | 279 | 277 | 183 | 176 | 60 | 64 | |||
Median PFS (95% CI) | 18.9 (15.2–21.4) mo | 10.2 (9.6–11.1) mo | 0.46 (0.37–0.57); p < .0001 | 15.2 (13.7–20.7) mo | 9.7 (8.4–11.1) mo | 0.44 (0.34–0.57); p < .0001 | 23.5 (17.8–24.3) mo | 15.0 (9.7–18.3) mo | 0.48 (0.28–0.80); p = .0047 |
AURA3 | |||||||||
Overall (n = 419) | Shedders (n = 272) | Nonshedders | |||||||
Osimertinib | Chemotherapy | HR (95% CI); p Value | Osimertinib | Chemotherapy | HR (95% CI); p Value | Osimertinib | Chemotherapy | HR (95% CI); p Value | |
Patients, n | 279 | 140 | 116 | 56 | NR | NR | |||
Median PFS (95% CI) | 10.1 (8.3–12.3) mo | 4.4 (4.2–5.6) mo | 0.30 (0.23–0.41); p < 0.0001 | 8.2 (6.8–97) mo | 4.2 (41.–5.1) mo | 0.42 (0.29–0.61) | NR | NR | NR |
AURA Phase I and AURA Extension Study (National Taiwan University Hospital study)32 | |||||||||
Shedder | Nonshedder | p Value | Shedder Without Brain Metastases | Nonshedder Without Brain Metastases | p Value | T790M “Drop Out” at Osimertinib Progression | Nonshedders at Osimertinib Progression | Shedders (New or Continual) at Osimertinib Progression | |
Patients, n | 40 | 13 | 23 | 12 | 11 | 12 | 17 | ||
Median PFS (during osimertinib treatment) | 9.4 (7.1–13.8) mo | 15.4 (9.6–NR) mo | .055 | 12.4 (8.4–20.9) mo | 15.4 (11.1–NR) mo | .26 | 2.6 (1.3–NR) mo | 11.1 (9.4–NR) mo | 12.6 (8.4–13.9) mo |
Median OS (during osimertinib treatment) | 13.5 (9.7–21.7) mo | 36.5 (33.3–NS) mo | .0157 | 19.8 (11.7–NR) mo | 36.5 (28.4–NR) mo | .089 | 9.3 (5.3–NA) mo | 22.4 (15.6–NA) mo | 15.4 (11.7–36.5) mo |
Patients, n | 38 | 9 | 21 | 8 | 11 | 12 | 17 | ||
Median survival after osimertinib progression | 5.0 (3.8–9.0) mo | 14.5 (7.2–NR) mo | .008 | 7.7 (5.0–NR) mo | 17.8 (12.2–NR) mo | .031 | 5.0 (3.0–NA) mo | 10.8 (7.2–NA) mo | 5.0 (3.1–16.5) mo |
Abbreviations: HR, hazard ratio; PFS, progression-free survival; NR, not reached; NA, not available; OS, overall survival.
Detection of RAS and BRAF Mutations in Colorectal Cancer
Retrospective analysis of two randomized phase III trials in metastatic colorectal cancer (mCRC; PRIME, CRYSTAL) comparing the addition of anti-EGFR antibody to chemotherapy versus chemotherapy alone indicated that multiple KRAS, NRAS, and BRAF mutations modulate the clinical response to anti-EGFR antibodies.35,36 In particular, an additional 14.7% (CRYSTAL) to 17% (PRIME) of non–exon 2 KRAS mutations were detected, which also did not benefit from EGFR monoclonal treatment, hence the detection of KRAS mutation should be extended to non–exon 2 KRAS mutations. The E.U. has approved three sets of liquid biopsy kits from two different companies that detect an extended spectrum of RAS mutations to select for patients with mCRC who will potentially benefit from anti-EGFR therapy and avoid the unnecessary expense and additional side effects of anti-EGFR antibody treatment.
OncoBEAM RAS CRC kit (KRAS and NRAS).
Several studies using different tissue comparator tests provided the evidence for approval of OncoBEAM RAS detection kit (Sysmex Inostics; Table 3) for the detection of KRAS and NRAS mutations in mCRC.37–42 Not surprisingly, when a much more sensitive tissue comparator test, such as NGS, is used, the sensitivity and specificity of BEAMing technology are not as high (e.g., with the Sanger sequencing method as the comparator; Table 3).43 The additional use of BEAMing to detect KRAS mutations includes real-time monitoring of treatment, detection of resistance mechanisms, and early detection of relapse and/or residual disease; these have been concisely reviewed by an expert task force panel.44
TABLE 3.
Performance Characteristics of OncoBEAM RAS CRC Kit, Idylla ctKRAS, and Idylla ctNRAS-BRAF Mutation Tests
OncoBEAM RAS CRC Kit | Idyllact KRAS Mutation Test | Idylla ctNRAS-BRAF Mutation Test | |
---|---|---|---|
CE Mark Approval Date | April 2016 | November 2017 | November 2017 |
Technology | BEAMing digital PCR | Idylla real-time PCR | Idylla real-time PCR |
Amount of Plasma Needed, mL | 3–4 | 1 | 1 |
Hands-on Time, min | Dedicated technician | < 1 | < 1 |
Turnaround Time | 1 wk | 130 min | 110 min |
Gene Coverage | 34 mutations: 16 KRAS and 18 NRAS | 21 KRAS | 18 NRAS |
Exon 2: codons 12 and 13 | Exon 2: codons 12 and 13 | Exon 2: codons 12 and 13 | |
Exon 3: codons 59 and 61 | Exon 3: codons 59 and 61 | Exon 3: codons 59 and 61 | |
Exon 4: codons 117 and 146 | Exon 4: codons 117 and 146 | Exon 4: codons 117 and 146 | |
5 BRAF | |||
Exon 15: codon 600 | |||
Sensitivity in Plasma, % | 0.02–0.04 | 0.5 | 0.5 |
MAF Quantification | Quantitative | Semiquantitative | Semiquantitative |
Clinical Validity Measures | OncoBEAM RAS CRC Kit, Instructions for | RASANC study (n = 198)45,46 | |
Use OBMRASIVD37 | Comparator test: plasma-based NGS analysis with sensitivity of 0.2% | ||
Tissue comparator test: standard of | KRAS+ (n = 84) | ||
care tissue testing | NRAS+ (n = 6) | ||
Sensitivity: 92.6% (112/121) | BRAF+ (n = 13) | ||
Specificity: 94.0% (110/117) | RAS. Sensitivity: 97.8% (90/92) | ||
Concordance: 93.3% (222/238) | RAS: Specificity: 95.3% (101/106) | ||
Grasselli et al, 2017 (n = 146)38 | RAS: Concordance: 96.5% (191/198) | ||
Tissue comparator test: PCR | KRAS: Concordance: 96% | ||
Sensitivity: 89% | NRAS: Concordance: 100% | ||
Specificity: 90% | BRAF: Concordance: 99.5% | ||
Concordance: 89.7% | |||
PPV: 84% | |||
NPV: 93% | |||
Vidal et al, 2017 (n = 115)39 | |||
Tissue comparator test: comparison: | |||
standard of care | |||
Sensitivity: 92.6% (112/121) | |||
Specificity: 94% (110/117) | |||
Concordance: 93.3% (222/238) | |||
Schmiegel et al, 2017 (n = 98)40 | |||
Tissue comparator test: Sanger | |||
sequencing | |||
Sensitivity: 90.4% (47/52) | |||
Specificity: 93.5% (43/46) | |||
Concordance: 91.8% (90/98) | |||
Normanno et al, 2017 (n = 92)43 | |||
Tissue comparator test: NGS | |||
Sensitivity: 69.7% | |||
Specificity: 83.1% | |||
Concordance: 78.3% (72/92) | |||
PPV: 69.7%; 83.1% | |||
Hahn et al, 2015 (n = 46)42 | |||
Comparator tumor test: Sanger | |||
sequencing or pyrosequencing | |||
Sensitivity: 92% (23/25) | |||
Specificity: 100% (21/21) | |||
Concordance: 96% (44/46) | |||
Clinical Outcome | Grasselli et al, 2017 (n = 146)38 | NR | |
Median OS: | |||
Tissue KRAS-wild-type: 39.1 mo | |||
Tissue KRAS-mutated: 28.7 mo | |||
Plasma KRAS-wild-type: 42.9 mo | |||
Plasma KRAS mutated: 27.8 mo |
Abbreviations: CE, Conformité Européene; PCR, polymerase chain reaction; MAF, mutant allele frequency; PPV, positive predictive value; NPV, negative predictive value; NGS, next-generation sequencing; NR, not reported.
Idylla ctKRAS mutation test and Idylla ctNRAS-BRAF mutation test.
The RASANC study provided the supporting evidence for the approval of the Idylla tests (Biocartis, Inc., Jersey City, NJ) for the detection of KRAS, NRAS, and BRAF in mCRC.45,46 Primary tumor removal, metachronous status, absence of liver metastases, and peritoneal carcinomatosis were significantly associated with mutant RAS tumor and negative plasma status, indicating the difference between shedders and nonshedders.45
In head and neck cancer, additional use of liquid biopsy to detect RAS mutations has been reported to identify resistance to cetuximab (anti-EGFR antibody)-based therapy.47 However, the resistance to anti-EGFR antibody therapy is not limited to RAS mutations based on liquid biopsy.48–50
BRAF V600E
BRAF V600E mutation is a validated and actionable driver mutation in melanoma and NSCLC.51,52 Surprisingly, to date there is little published literature on the clinical use of liquid biopsy to detect BRAF V600E mutation in melanoma and NSCLC.53 Most of the literature on liquid biopsy for BRAF V600E is in mCRC, especially when used in extended-spectrum RAS mutational analysis to select patients for anti-EGFR treatment (Table 3). Not surprisingly, a greater amount of BRAF V600E detected at liquid biopsy was associated with worse prognosis for survival in mCRC (hazard ratio [HR], 7.33; 95% CI, 1.04–2.89; p = .002) in a multivariate analysis that included age, tumor location, carcinoembryonic anti-gen (CEA), KRAS mutational status, and amount of circulating cell-free DNA.54
PIK3CA Mutations
Although no approved drug specifically targets PIK3CA mutations, and PIK3CA mutations have not been conclusively shown to be a driver mutation, PIK3CA mutations may confer sensitivity to PIK3CA inhibitors. Two randomized phase III trials have investigated the potential clinical benefit of the addition of buparlisib (BKM210), a class 1 pan-PIK3CA inhibitor, to fulvestrant in hormone receptor–positive, HER2-negative patients whose disease is refractory to aromatase inhibitors (AIs; BELLE-2) and additionally refractory to mTOR inhibition (BELLE-3).55,56 Both trials investigated the presence of plasma PIK3CA mutations, established by using BEAMing technology, and its implication for clinical outcome. Overall, the most common PIK3CA mutations identified from ctDNA from BELLE-3 were H1047R (39.7%), E545K (36.8%), E542K (18.4%), and H1047L (5.1%).
The sensitivity of the plasma PIK3CA mutations compared with tissue PIK3CA mutations in BELLE-2 was 71.2% (99/139), specificity was 79.2% (244/307), and overall concordance was 76.7% (342/446). Similarly, for BELLE-3, sensitivity was 80% (70/87), specificity was 87% (142/163), and concordance was 84.8% (212/250). The lower concordance in BELLE-2 probably results from the fact that detection of PIK3CA mutation in tumor is performed by using Sanger sequencing, which is not as sensitive as BEAMing technology. The presence of PIK3CA mutations in plasma corresponded to improved PFS in patients who received buparlisib in addition to fulvestrant compared with patients who received only fulvestrant in both BELLE-2 and BELLE-3 trials (Table 4).55,56 However, neither trial analyzed the prognostic significance of shedders versus nonshedders of PIK3CA mutations.
TABLE 4.
Clinical Outcomes of BELLE-2 and BELLE-3 According to PKI3CA Mutation Status in Tumor and Plasma
Variable | Overall Population | ctDNA PIK3CA+ | ctDNA PIK3CA− | Tumor PIK3CA+ | Tumor PIK3CA− | |||||
---|---|---|---|---|---|---|---|---|---|---|
Buparlisib and Fulvestrant | Placebo and Fulvestrant | Buparlisib and Fulvestrant | Placebo and Fulvestrant | Buparlisib and Fulvestrant | Placebo and Fulvestrant | Buparlisib and Fulvestrant | Placebo and Fulvestrant | Buparlisib and Fulvestrant | Placebo and Fulvestrant | |
BELLE-2* | ||||||||||
Patients, n | 576 | 571 | 87 | 113 | 199 | 188 | 21 | 19 | 123 | 120 |
ORR, % (95% CI) | 11.8 (9.3–14.7) | 7.7% (5.7–10.2) | 18.4 (10.9–28.1) | 3.5 (1.0–8.8) | 11.6 (7.5–16.8) | 10.6% (6.6–16.0) | NR | NR | NR | NR |
Median PFS, mo (95% CI) | 6.9 (6.8–7.8) | 5.0 (4.0–5.2) | 7.0 (5.0–10.0) | 3.2 (2.0–5.1) | 6.8 (4.7–8.5) | 6.8 (4.7–8.6) | 4.4 (1.6–NE) | 10.7 (3.0–NE) | 5.1 (3.5–8.5) | 4.7 (3.3–8.5) |
HR (95% CI) | 0.78 (0.67–0.89) | 0.58 (0.41–0.82) | 1.02 (0.79–1.30) | 1.18 (0.49–2.85) | 0.98 (0.72–1.32) | |||||
p Value | .00021 (1-sided) | .001 (1-sided) | .557 (1-sided) | NR | NR | |||||
BELLE-3** | ||||||||||
Patients, n | 289 | 143 | 100 | 35 | 132 | 81 | 75 | 34 | 135 | 69 |
ORR, % (95% CI) | 3.9 (2.8–4.2) | 1.8 (1.5–2.8) | 4.2 (2.8–6.7) | 1.6 (1.4–2.8) | 3.9 (2.8–4.3) | 2.7 (1.5–3.6) | 4.7 (2.9–6.7) | 1.4 (1.4–2.2) | 2.8 (1.9–3.4) | 1.7 (1.4–2.9) |
Median PFS, mo (95% CI) | 0.67 (0.53–0.84) | 0.46 (0.29–0.73) | 0.73 (0.53–1.00) | 0.39 (0.23–0.65) | 0.81 (0.59–1.12) | |||||
HR (95% CI) | .0030 | .0031 | .026 | < .0001 | .099 |
BELLE-2: Women who were estrogen receptor–positive or progesterone receptor–positive, human epidermal growth factor receptor 2–negative, aromatase inhibitor refractory. ctDNA PKI3CA determined by BEAMing polymerase chain reaction; tumor PIK3CA determined by Sanger sequencing.
BELLE-3: Women who were estrogen receptor-positive or progesterone receptor–positive, human epidermal growth factor receptor 2–negative, aromatase inhibitor refractory, and refractory to mTOR inhibition. ctDNA PIK3CA determined by Inostics BEAMing polymerase chain reaction, covering exons 9 and 20; tumor PIK3CA determined by Cobas PIK3CA assay covering exons 7, 9, and 20.
Abbreviations: ORR, overall response rate; NR, not reported; NE, not evaluable; PFS, progression-free survival; HR, hazard ratio.
The role of PIK3CA mutation in breast cancer is being further evaluated in two phase III trials: SOLAR-1 () is comparing alpelisib (a PI3KA inhibitor) and fulvestrant versus fulvestrant alone, with a prospective analysis of PIK3CA mutations in ctDNA, and SANDPIPER () is comparing taselisib (GDC-0032) and fulvestrant versus fulvestrant alone in PIK3CA-mutant breast cancer as determined by Cobas testing. Otherwise, the eligibility criteria for both trials are similar to those of BELLE-2 (disease that is refractory to AI). If these trials will lead to the eventual approval of PIK3CA inhibitors, then PIK3CA mutations detection will become very important.
The prognostic significance of pretreatment plasma PIK-3CA in cervical cancer was also investigated in 117 patients with cervical cancer treated in Hong Kong over a period of 10 years (1997 to 2007). ddPCR was used to detect PIK3CA E542K and E545K mutations.57 Overall, 26 of 117 (22.2%) patients were identified (23.7% in squamous cell carcinoma of the cervix and 15.0% in adenocarcinoma of the cervix). The presence of plasma PIK3CA mutations was associated with high pathologic grade and large tumor size (mean tumor size, 4 cm) compared with 3 cm for patients negative for plasma PIK3CA mutations but was not associated with age, clinical stage, presence or absence of lymphovascular invasion, or pelvic lymph node metastasis. Although PIK3CA mutation was associated with shortened disease-free survival and OS on univariate analysis, on multivariate analysis PIK3CA mutation did not affect disease-free survival (HR 1.449; p = .464) or OS (HR 1.261; p = .643). Going forward, in analyses of larger cohorts of patients with cervical cancer who have uniform stage, histology, histologic grade, and treatment modality and are from a more contemporaneous treatment period, plasma PIK3CA mutations may turn out to be an important prognostic factor in cervical cancer. Liquid biopsy for PIK3CA mutation has also been investigated in small numbers of patients with bladder cancer and mCRC.58,59
HER2 Amplification
HER2 amplification was validated as an actionable target in breast cancer in 2001 and in gastric adenocarcinoma in 2010.60,61 The gold standard of detecting HER2 amplification in breast cancer is by immunohistochemistry or florescence in situ hybridization. Given the generally ready availability of tumor tissue from patients with breast cancer, there is scant literature on the use of liquid biopsy to detect or to monitor HER2 amplification at diagnosis or during treatment, respectively. Small studies have demonstrated that HER2 amplification detected in plasma correlates well with tumor HER2 amplification and that the level of HER2 amplification can be used to monitor treatment and recurrence in gastric cancer.62,63 HER2 amplification was successfully identified by using a liquid biopsy NGS panel to identify driver mutations to enroll in a basket targeted therapy trial that included HER2 inhibition.64
USE OF LIQUID BIOPSY TO DETERMINE RESISTANCE MECHANISMS TO HORMONAL BLOCKADE IN SOLID TUMORS
Estrogen Receptor (ESR1) Mutation
Detection of ESR1 mutation (primarily in the ligand-binding domain) in plasma has been investigated as a resistance mechanism to an AI in two randomized phase III trials, SoFEA (Study of Faslodex Versus Exemestane With or Without Arimidex) and PALOMA3 (Palbociclib Combined With Fulvestrant in Hormone Receptor–Positive HER2-Negative Metastatic Breast Cancer After Endocrine Failure), using the Bio-Rad QX-200 ddPCR system. In the SoFEA analysis, patients with mutant ESR1 in the exemestane treatment arm had significantly worse PFS (p = .01) than those without mutated ESR1. Similarly, PFS was significantly improved (p = .02) in patients with mutant ESR1 when they received fulvestrant-containing regimens but not (p = .77) in patients with wild-type ESR1 (Table 5). On the other hand, not surprisingly, the retrospective analysis of the PALOMA3 showed that both ESR1-mutated (HR 0.43; 95% CI, 0.25–0.74; p = .002) and ESR1 wild-type (HR 0.49; 95% CI, 0.35–0.70; p < .001) patients benefited from the addition of palbociclib to fulvestrant compared with those receiving placebo and fulvestrant because palbociclib is a CDK4/6 inhibitor (Table 6).65
Table 5.
Effect of ESR1 Mutations on Clinical Outcome in Patients Receiving Endocrine Therapies in SoFEA Trial
Variable | Exemestane | Hazard Ratio (95% CI); p Value | |
---|---|---|---|
ESR1 Wild-Type | ESR1-Mutated | ||
Patients (n) | 39 | 18 | |
Median PFS, mo (95% CI) | 8.0 (3.0–11.5) | 2.6 (2.4–6.2) | 2.12 (1.18–3.81); .01 |
Median OS, mo (95% CI) | 22.8 (17.6–32.4) | 12.8 (5.7–27.0) | 1.65 (0.81–3.38); .16 |
ESR1-Mutated | |||
Fulvestrant Regimens | Exemestane | ||
Patients (n) | 45 | 18 | |
Median PFS, mo (95% CI) | 5.7 (3.0–8.5) | 2.6 (2.4–6.2) | 0.52 (CI, 0.30–0.92); .02 |
ESR1 Wild-Type | |||
Fulvestrant Regimens | Exemestane | ||
Patients (n) | 59 | 39 | |
Median PFS, mo (95% CI) | 8.0 (3.0–11.5) | 5.4 (3.7–81.) | 1.07 (0.68–1.67); .77 |
Abbreviation: PFS, progression-free survival.
Table 6.
Effect of ESR1 Mutations on Clinical Outcome in Patients Receiving Endocrine Therapies in PALOMA3 Trial
Variable | ESR1-Mutated | Odds Ratio or Hazard Ratio (95% CI); p Value Statistical Consideration | |
---|---|---|---|
Palbociclib + Fulvestrant | Placebo + Fulvestrant | ||
Patients, n | 63 | 28 | |
ORR, % | 17.5 | 3.6 | 5.94* (0.78–270.5); .06 |
Median PFS, mo (95% CI) | 9.4 (5.3–11.1) | 3.6 (2.0–5.5) | 0.43** (0.25–0.74); .002 |
ESR1 Wild Type | |||
Palbociclib + Fulvestrant | Placebo + Fulvestrant | ||
Patients, n | 177 | 92 | |
ORR,% | 21.5 | 14.1 | 1.66* (0.81–3.61); .14 |
Median PFS, mo (95% CI) | 9.5 (9.2–NE) | 5.4 (3.5–7.4) | 0.49** (0.35–0.70); ≤.001 |
ESR1 Status Available | |||
ESR1 Wild Type | ESR1-Mutated | ||
Patients, n | 261 | 91 | |
Median PFS (95% CI) | 9.2 (7.4–10.9) | 7.3 (3.7–9.1) | 1.46** (1.06–2.02); .02 |
Odds ratio (univariate analysis).
Hazard ratio (univariate analysis).
Abbreviations: ORR, overall response rate; NE, not evaluable.
Together, these two trials suggest that for patients who progressed while receiving a nonsteroidal AI and have detectable ESR1 mutations, it is best to switch to fulvestrant-containing regimens, such as fulvestrant and palbociclib. For patients who progressed while receiving a nonsteroidal AI and have no detectable ESR1 mutations, switching to an irreversible AI, such as exemestane, remains a treatment option before proceeding to fulvestrant-contain regimens. These are hypothesis-generating observations because only 161 of 723 (22.3%) patients enrolled in the SoFEA and only 360 of 551 (65.3%) patients enrolled in PALOMA3 had plasma available and successfully analyzed.
In the PALOMA3 trial, logistic multivariate analysis showed that the presence of plasma ESR1 mutations was negatively associated with tamoxifen treatment only (odds ratio [OR], 0.06; 95% CI, 0.01–0.45; p = .01) and positively associated with sensitivity to endocrine therapy (OR, 3.95; 95% CI, 1.59–9.78; p = .003), bone metastasis (OR, 3.31; 95% CI, 1.54–6.39; p = .002), and visceral metastasis (OR, 1.74; 95% CI, 1.02–2.98; p = .04). Importantly, multivariate analysis of the PFS benefit in PALOMA3 showed a negative association with plasma ESR1-mutated genotype (HR 1.4; 95% CI, 1.07–2.08; p = .02), even after factoring in the significant benefit from palbociclib.65
Recent analysis of a large trial of first-line AI therapy in breast cancer indicated that ESR1 mutations may play a role in AI resistance, but other resistance mechanism, such as KRAS, have been identified.66 Finally, liquid biopsy using NGS indicated multiple resistance mechanisms to AI in addition to ESR1 mutations.67
Androgen Receptor Variant 7
A splice variant of the androgen receptor variant 7 (AR-V7) that deletes the ligand domain of AR isolated from circulating tumor cells in the plasma is involved in de novo and acquired resistance to abiraterone (an androgen synthesis inhibitor) and enzalutamide (an AR ligand–binding domain antagonist), both of which have been approved for the treatment of castration-resistant prostate cancer (Table 7). In multivariable Cox regression analysis stratified by treatment type, AR-V7 detection remained independently predictive of prostate-specific antigen–defined PFS (HR 8.2; 95% CI, 2.7–24.9; p < .001), clinical/radiographic PFS (HR 4.9; 95% CI, 1.7–13.8; p = .003), or OS (HR 5.0; 95% CI, 1.3–19.8; p = .021; Table 7).68 Further study has indicated that AR amplification was associated with resistance to enzalutamide, and mutations (H874Y, T877A, D879E, L881I, E893K, and M895V) in the ligand-binding domain of AR are more associated with resistance to abiteratone.69 More recently, a longitudinal multiplex targeting sequencing of plasma from a study of 65 patients with castration-resistant prostate cancer receiving enzalutamide indicated that besides AR amplification of AR mutations, RB1 loss (p = .01) and MET copy number gain/amplification (p = .2) were significantly associated with shorter PFS after adjustment for the presence of ctDNA.70 Overall mutations in the direct target of hormonal blockade can partially explain the resistance to hormonal treatment of breast and prostate cancer. However, the advent of NGS liquid biopsy has allowed the discovery a much more complex pattern of resistance mechanisms.
TABLE 7.
Androgen Receptor Variant 7 Detected by Quantitative Reverse Transcriptase Polymerase Chain Reaction in Circulating Tumor Cells as Both Primary and Acquired Resistance to Abiraterone and Enzalutamide
Abiraterone (n = 31) | Enzalutamide (n = 31) | |||
---|---|---|---|---|
Pretreatment AR-V7–Positive | Pretreatment AR-V7–Negative | Pretreatment AR-V7–Positive | Pretreatment AR-V7–Negative | |
Patients, n | 6 | 25 | 12 | 19 |
PSA ORR, % (95% CI) | 0 (0–46) | 68 (46–85) | 0 (0–26) | 53 (29–76) |
p Value | .004 | .004 | ||
PSA PFS, mo (95% CI) | 1.3 (0.9–NR) | 5.3 (5.3–NR) | 1.4 (0.9–NR) | 6.0 (3.8–NR) |
HR (95% CI) | 16.1 (3.9–66.0) | 7.4 (2.7–20.6) | ||
p Value | < .001 | < .001 | ||
PFS*, mo (95% CI) | 2.3 (1.4–NR) | 6.3 (6.3–NR) | 2.1 (2.0–NR) | 6.1 (4.7–NR) |
HR (95% CI) | 16.5 (3.3–82.9) | 8.5 (2.8–25.5) | ||
p Value | < .001 | < .001 | ||
OS, mo | 10.6 | NR | 5.5 | NR |
HR (95% CI) | 12.7 (1.3–125.3) | 6.9 (1.7–28.1) | ||
p Value | .006 | .002 | ||
Pretreatment (n = 62) | Pretreatment AR-V7–Negative (n = 42) | |||
AR-V7–Positive | AR-V7–Negative | Post-Treatment AR-V7–Negative | Post-Treatment AR-V7–Positive | |
Patients, n | 18 | 44 | 36 | 6 |
ORR, % (95% CI) | 0 (0–19) | 61 (45–76) | 68 (52–81) | 17 (4–58) |
p Value | < .0001 | NA | ||
PSA PFS, mo (95% CI) | 1.4 (0.9–2.6) | 6.1 (5.3–NR) | 6.1 (5.9–NA) | 3.0 (2.2–NA) |
HR (95% CI) | 10.5 (4.7–23.6) | NA | ||
p Value | < .001 | NA | ||
PFS*, mo (95% CI) | 2.1 (1.9–3) | 6.4 (6.1–NR) | 6.5 (6.1–NA) | 3.2 (3.1–NA) |
HR (95% CI) | 12.7 (5.1–31.9) | NA | ||
p Value | < .001 | NA | ||
OS (95% CI) | 9.2 (4.5–NR) | >11.9 (11.9–NR) | NA | NA |
HR (95% CI) | 8.3 (2.5–27.4) | NA | ||
p Value | < .001 | NA |
Clinical or radiographic; PSA response is defined as ≥50% decline in baseline PSA maintained for ≥4 weeks.
Abbreviations: AR-V7, androgen receptor-variant 7; PSA, prostate-specific antigen; ORR, overall response rate; PFS, progression-free survival; NR, not reached; HR, hazard ratio; OS, overall survival; NA, not available.
NEXT-GENERATION SEQUENCING PLATFORMS
Liquid biopsy using NGS will be the dominant platform, with its increased depth of coverage, simultaneous multigene sequencing, and ability to simultaneously detect all modes of genomic alterations (point mutations, insertion/deletion [indel], amplification, and gene rearrangements) to account for both tumor heterogeneity and multiple resistance mechanisms and determination of tumor mutation burden (TMB). The two most common commercially used liquid biopsies are Guardant360 (Guardant Health, Redwood City, CA) and FoundationACT (Foundation Medicine Inc., Cambridge, MA). As listed in Table 8, the limit of detection and the gene panel of Guardant360 has evolved over time. It is important to be aware of the number of exact genes included in the NGS sequencing platform by consulting with the company websites because technology advances will lead to increased complexities of the NGS panel being offered commercially. It is also important to note how the interpretation of the literature depends on the various versions of Guardant360.
TABLE 8.
Comparison of Various and Most Current Versions of Guardant360 and Foundation Medicine FoundationACT Liquid Biopsy Platforms (as of December 31, 2017)
Guardant360 54-Gene Panel | Guardant360 68-Gene Panel | Guardant360 70-Gene Panel | Guardant360 73-Gene Panel*71 | Foundation Medicine FoundationACT 62-Gene Panel*72 | |
---|---|---|---|---|---|
Commercial Launch Date | June 2014 | August 2015 | October 2015 | November 2016 | June 2016 |
Amount of Blood Needed | Two 10-mL Streck tubes | Two 10-mL Streck tubes | Two 10-mL Streck tubes | Two 10-mL Streck tubes | Two 10-mL tubes (Foundation ACT Specimen Collection Kit) |
Minimal Amount of Cell-Free DNA | > 5 ng of cell-free DNA1 | > 5 ng of cell-free DNA1 | > 5 ng of cell-free DNA1 | > 5 ng of cell-free DNA1 | ≥ 20 ng of cell-free DNA |
Methods for Isolating ctDNA | QIAamp circulating nucleic acid kit (Qiagen) | QIAamp circulating nucleic acid kit (Qiagen) | QIAamp circulating nucleic acid kit (Qiagen) | QIAamp circulating nucleic acid kit (Qiagen) | |
Types of Alterations Analyzed and No. of Genes | Complete exons (18 genes) | Complete exons (29 genes) | Complete exons (30 genes) | Complete exons (19 genes)** | Complete exons (27 genes) |
Critical exons (36 genes) | Critical exons (39 genes) | Critical exons (40 genes) | Critical exons (54 genes) | Critical exons (33 genes) | |
CNV (3 genes) | Indel (1 gene) | Indels (3 genes) | Indels (23 genes) | Indel (27genes) | |
CNV (16 genes) | CNV (18 genes) | CNV (18 genes) | Amplification (27 genes) | ||
Fusions (4 genes) | Fusions (6 genes)† | Fusions (6 genes)† | Fusions (6 genes)†‡ Promoter region only (TERT) |
||
Sequencing Platform | HiSEquation 2500 (lllumina)l, 2 | HiSeq 2500 or NextSEquation 500 (lllumina) | HiSeq 2500 or NextSEquation 500 (lllumina) | HiSeq 2500 or NextSEquation 500 (lllumina) | HiSeq 2500 or HiSeq 4000 (lllumina) |
Depth of Coverage | ?10,000× (average raw) | ~15,000×(average raw) | 7,503× (median, unique, actual depth of coverage reported) | ||
Single-Nucleotide Variation | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | ≥ 0.04%; ≥ 0.25%, sensivitiy 100%, PPV 99.2%; 0.05–0.25% sensitivity 63.8%, PPV 96.3% | ≥ 0.1% (0.1%–0.5%; sensitivity, 67.3%;PPV, 93.6%) ≥ 0.05% (sensitivity > 98.9%; PPV > 99.9%) |
Indel | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | ≥ 0.02%; ≥ 0.02%, sensitivity 100%, PPV 98.2%; 0.05–0.2% sensitivity 67.8%, PPV 98.2% | ≥ 1% (sensitivity, 99.9%; PPV, 98.8%) |
CNV/Amplification | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | ≥ 2.12 copies | ≥ 20% (sensitivity, 95.3%; PPV, 97.5%) |
Fusions/Rearrangement | Varies between < 0.1% and 0.1%; depends on particular gene | Varies between < 0.1% and 0.1%; depends on particular gene | ≥ 0.04%; ≥ 0.2% sensitivity 95%, PPV 100%; 0.05–0.2% sensitivity 83%, PPV 100% | ≥ 1% (sensitivity > 99.9%; PPV, 98.8%) < 1% (sensitivity, 86.8%; PPV > 99%) |
|
Turnaround Time for Results | Within 2 wk of receiving blood sample | Median of 7 calendar days of receiving blood sample | Within 2 wk of receiving blood sample | ||
Difference in Gene Panel | ABL, CSF1R, ERBB4, FLT3, KDR (VEGFR2), PROC, RB1, and SMARCB1 genes were deleted and replaced with ARAF, ARID1A, BRCA1, BRCA2, CCND1, CCND2, CCNE1, CDK4, CDK6, CDKN2B, ESR1, GATA3, MAP2K1, MAP2K2, NF1, NTRK1, NTRK3, NFE2L2, RHEB, RHOA, RIT1, RAF1, and ROS1 genes. Copy number amplification calling increased from 3 to 16 genes (added AR, BRAF, CCND1, CCND2, CCNE1, CDK4, CDK6, FGFR1, FGFR2, KIT, KRAS, MYC, PDGFRA, PIK3CA, RAF1J Fusing calling added for 4 genes (ALK, ROS1, RET, NTRK1). Indel calling added for EGFR (exon 19/20) | RB1 and TSC1 genes were added; number of fusions detected increased from 4 to 6 (additional FGFR2 and FGFR3 fusion detection). Indel coverage for ERBB2 (exon 19/20) and MET exon 14 splice site variants wasadded | SRC and CDKN2B were deleted from panel and DDR2, MAP-K1(ERK2), MAPK3 (ERK1), MTOR, and NTRK3 were added. Copy number amplification calling was increased to 18 genes (CCND1, CCND2). Indel coverage expanded from 3 to 23 genes (ATM, APC, ARID1A,BRCA1, BRCA2, CDH1,CDKN2A, GATA3, KIT, MLH1,MTOR, NF1, PDGFRA, PTEN, RB1,SM AD4,ST-K11,TP53,TSC1,VHL) | NA | |
Cost | NA | NA | NA | $5,800 per test ($5,000 suggested contracted price per test) | $5,800 USD per test (contracted price may vary) |
References (Selected Clinical Use of One Particular Platform) | 73–76 | 75–79 | 64,75,76,80,81 | 82,83 | 84–87 |
Although the difference in the number of genes is 11 between the current Guardant360 and Foundation Medicine FoundationACT, the actual difference in the number of genes analyzed is 31 because of unique genes analyzed by each liquid biopsy. Twenty-one genes were unique to Guardant360: AR, ARID1A, ATM, FBXW7, GATA3, HNF1A, MAPK3, MLH1, MPL, NFE2L2, NOTCH1, NTRK1, NTRK3, RB1, RHEB, THOA, RIT1, SMAD, STK11, TSC1, VHL. Ten genes were unique to Foundation Medicine FoundationACT: ABL, BTK, CRKL, ERRFI1, FOXL2, MDM2, MYD88, PDGFR-beta, PD-L2, VEGF-alpha.
The 19 genes that have complete exon coverage in the Guardant 73-gene panel are as follows: APC, AR, BRAF, BRAC1, BRCA2, EGFR, ERBB2, HRAS, KRAS, NRAS, KIT, MAPK1(ERK2), MAPK2 (ERK1), MET, MYC, PIK3CA, RB1, TP53, STK11. Underlined genes have complete exon coverage by Foundation Medicine FoundationACT.
Both Guardant and Foundation Medicine FoundationACT report four common fusions: ALK, ROS1, RET, and FGFR3 fusions. Guardant360 also reports additional NTRK1 and FGFR2 fusions, and Foundation Medicine FoundationACT reports additional EGFR and PDGFR-alpha fusions.
ROS1 rearrangement is the only ROS1 alteration detected by the current version of Foundation Medicine FoundationACT.
Abbreviations: PPV, positive predictive value; NA, not available; USD, US dollars.
Among the many other NGS liquid biopsy platforms that are available or under development are Oncomine Lung cfDNA Assay (ThermoFisher Scientific, Waltham, MA),88 Archer Reveal ctDNA (ArcherDX, Boulder, CO),89 Oncotype SEQ (Genomic Health, Redwood City, CA),90 LiquidDx (MolecularMD, Portland, OR),91 CancerIntercept Detect and CancerIntercept Monitor (Pathway Genomics, San Diego, CA),92 OptiSeq NGS Pan-Cancer Panel (DiCarta, Richmond, CA),93 and PlasmaSELECT (Personal Genome Diagnostics, Baltimore, MD).94
USE OF LIQUID BIOPSY TO DETERMINE TUMOR MUTATION BURDEN
TMB is usually expressed as number of mutations per mega-base of DNA (MB)95 and has been shown to be predictive to response to anti–PD-L1 therapy.96 In the CheckMate026 trial, which compared nivolumab to platinum-based chemotherapy as first-line treatment of NSCLC, OS did not differ between nivolumab and chemotherapy. A subgroup analyzed with TMB, as determined by whole-exome sequencing, indicated that patients with NSCLC and high TMB had a marginally significantly improved PFS compared with those receiving platinum-based chemotherapy (9.7 months [95% CI, 5.1 months to not reached] versus 5.8 months [95% CI, 4.2–8.5 months]; HR 0.62 [95% CI, 0.38–1.00]). OS did not differ between nivolumab and chemotherapy regardless of high or low TMB.96 Importantly, there is excellent concordance between TMB as determined by a commercially available (Foundation Medicine Inc.) tissue-based hybrid-capture NGS method and whole-exome sequencing of the tumor but essentially no correlation between TMB and PD-L1 expression.96
It is now feasible to determine TMB by liquid biopsy using whole-exome sequencing or NGS hybrid-capture method.97,98 The blood TMB (bTMB) assay developed by Foundation Medicine Inc. uses 10 mL of plasma to deliver an account of somatic base substitutions down to 0.5% allele frequency across 394 genes from as little as 1% tumor content in a cell-free DNA sample. Computational methods differ: bTMB analyzes only single-nucleotide variants (SNVs), whereas tissue TMB also includes analysis of indels and fusions. Sensitivity, specificity, and positive predictive value were 93.9%, 100.0%, and 100.0%, respectively, when compared with tissue TMB.98 Importantly, the bTMB assay demonstrates strong sensitivity and specificity between the cut-points of 8–20 mutations/MB. The clinical utility of the bTMB assay was retrospectively analyzed in 783 patients with NSCLC receiving second-line therapy (OAK and POPLAR); PFS and OS were compared between atezolizumab (an anti–PD-L1 antibody) and single-agent docetaxel as second-line treatment. At a bTMB cutoff of 16 or greater, PFS and OS significantly improved in the phase II randomized POPLAR study.99 Among the 850 patients enrolled in the phase III OAK trial, 583 patients were considered as the “biomarkers evaluable population” (BEP) for bTMB. A bTMB of 16 or greater represented 27% of the BEP. There was an OS benefit for patients with both bTMB of 16 or greater (HR 0.64; 95% CI, 0.44–0.92) and bTMB less than 16 (HR 0.65; 95% CI, 0.52–0.81). Indeed, PFS (HR 0.73; 95% CI, 0.56–0.95) and OS (HR 0.69; 95% CI, 0.52–0.93) benefits were observed in patients with bTMB of 10 or less. A bTMB of 10 or less represents 43% of the BEP, and the sensitivity and specificity were both 100% when compared with tissue TMB.99 The bTMB assay is being validated in two trials. B-F1RST () is a phase II single-arm study of atezolizumab as first-line treatment of NSCLC while prospectively collecting plasma to evaluate whether bTMB can predict for improved clinical outcome with atezolizumab. B-FAST () is prospectively enrolling patients with NSCLC receiving first-line therapy in a global, multicenter, multicohort, randomized phase II/III trial without certain targetable driver mutations (RET or ALK fusion) detected based on blood-based assays; additionally, the trial is comparing single-agent atezolizumab therapy to standard of care based on bTMB results for those patients without RET or ALK rearrangement.100
USE OF CTDNA IN LIQUID BIOPSY TO DETECT MINIMAL RESIDUAL DISEASE IN SOLID TUMORS
Most solid tumors do not have a single truncal actionable driver mutation. There is vast literature on the use of nonspecific ctDNA in solid malignancies ranging from detection to monitor treatment response to detection of recurrence, and even for survival prognostication, all of which are beyond the scope of this review. However, ctDNA detection by liquid NGS has been used to identify minimal residual disease (MRD); we review two important studies here.101,102
In a study of 230 patients with resected stage II colon cancer conducted by Tie and colleagues, 20 (8.7%) patients had ctDNA detected by massive parallel sequencing. Most of these mutations were TP53, APC, and KRAS. There was no difference in clinicopathologic difference (age, sex, right-versus left-sided colon, tumor differentiation, T3 versus T4, number of lymph node resected < 12 versus ≥ 12, mismatch repair status, presence or absence of lymphovascular invasion, with or without adjuvant chemotherapy) between patients with and without detectable ctDNA. Among the 178 patients not treated with adjuvant chemotherapy, postoperative recurrence occurred in 11 of 14 (78.6%) ctDNA-positive patients compared with 16 of 164 (9.8%) ctDNA-negative patients. The 3-year relapse-free survival rate was 0% for ctDNA-positive patients compared with 90% for ctDNA-negative patients (HR 18; 95% CI, 7.9–40; p = 2.5 × 10−12). By multivariate analysis, ctDNA positivity (HR 28; 95% CI, 11–68; p = .0001) and T stage (T4 versus T3; HR 8.1; 95% CI, 3.1–21; p < .0001) were the two independent predictive factor for RFS after factoring in the above clinicopathologic factors. In addition, among the 230 patients, ctDNA positivity (HR 14; 95% CI, 6.8–28; p < .0001) and T stage (HR 2.6; 95% CI, 3.1–5.5; p = .001) remained the two independent factors for PFS after factoring in adjuvant chemotherapy. More importantly, postoperative elevation of CEA level was not a predictive factor for relapse-free survival, even by univariate analysis with or without adjuvant chemotherapy.101 Furthermore, ctDNA positivity immediately after completion of postoperative chemotherapy was significantly associated with poor relapse-free survival (HR 11; 95% CI, 1.8–6.8; p = .001). However, the number of patients analyzed was 44, and only three patients had ctDNA positivity immediately after completion of postoperative chemotherapy.
Finally, during the follow-up period, ctDNA was significantly more frequently positive (23/27 [85%]) than was CEA elevation (11/27 [41%]) at the time of radiographic recurrence (p = .002). The time between ctDNA positivity (median, 167 days) was significantly longer than that between CEA elevation (median, 61 days) to radiographic recurrence (p = .04). This retrospective analysis of prospective collected data indicated that ctDNA monitoring has the ability to detect MRD and hence early recurrence, leading to the potential initiation and/or lengthening of adjuvant chemotherapy based on the presence or absence of ctDNAs. There is a randomzied ctDNA driven adjuvant chemotherapy trial in resected stage II CRC in Australia/New Zealand (DYNAMIC, Trial ID number: ACTRN12615000381583). In the standard-of-care arm, adjuvant chemotherapy wil be given as determined by the clinicians, while in the investigational arm, adjuvant chemothrapy will be guided by the presence (adjuvant chemotherapy) or absence (no adjuvant chemotheray) of postoperative ctDNA in resected stage II CRC. The primary endpoint is to evaluate whether an adjuvant therapy strategy based on ctDNA results may affect the number of patients treated with chemotherapy and recurrence-free survival.
TRACERx is a U.K. study that developed proprietary NGS technology to link the individual patient’s tumor DNA to plasma ctDNA by identifying two distinct tumor-associated mutations (usually SNV).102 The resected tumor was further microdissected at several different areas, and all “subtumors” underwent NGS sequencing to detect tumor heterogeneity.103 The TRACERx study group used this phylogenetic ctDNA (at least two SNVs) tracking to detect recurrence in 96 patients with resected early-stage lung cancer. A group of 24 patients were followed longitudinally with pre- and postoperative plasma ctDNA; of these, 10 had been relapse free for a median of 775 days, whereas 14 had confirmed relapse. Thirteen of the 14 patients who relapsed had ctDNA detected before or at clinical relapse compared with only one of 10 patients in remission (two SNVs). Overall, the median duration between ctDNA detection and radiographic evidence of relapse was 70 days. Relapse can involve one or more subclones. This analysis has some limitations due to lack of regularly scheduled imaging to account for potential lead-time bias of regular liquid biopsy compared with detection of recurrence by clinical practice.104 Regardless, the TRACERx approach remains an important technology that accounts for tumor heterogeneity in the primary tumor, especially because most lung cancers do not harbor a signature actionable driver alteration.
Detection of MRD may turn out to be the most important aspect of use of liquid biopsy because it may help guide extent of adjuvant treatment,105 similar to the use of Epstein-Barr virus load in the treatment of early-stage nasopharyngeal carcinoma.106
SHEDDERS VERSUS NONSHEDDERS OF ACTIONABLE GENOMIC ALTERATIONS
In the TRACERx study, 46 of 96 patients had at least two SNVs that allowed phylogenetic ctDNA tracking, including 30 of 31 (97%) lung squamous cell carcinomas, 11 of 58 (19%) lung adenocarcinomas, and five of seven other NSCLCs. Among stage I NSCLC, 16 of 17 squamous cell carcinomas (94%) compared with five of 39 (13%) adenocarcinomas had detectable ctDNA. Of note, the median necrosis rate in lung squamous cell carcinoma (40%; n = 31) was significantly higher than in ctDNA-positive lung adenocarcinomas (15%; n = 11) and ctDNA-negative adenocarcinomas (2%; n = 47; p < .0001). Multivariate analysis indicated that nonadenocarcinoma histology (p = .001), lymphovascular invasion (p = .042), and high Ki-67 proliferation index (p = .022) are independent predictors of ctDNA positivity but not tumor necrosis (10% increment increase; p = .862), tumor size (1-cm increment increase; p = .134), or amount of cfDNA (p = .229). Importantly, the mean plasma variant allele frequency positively corresponded to tumor size. The TRACERx team was able to estimate that a mean clonal plasma variant allele frequency of 0.1% corresponds to a tumor burden of 10 cm3 or a burden of 302 million tumor cells.102
A large-scale systemic review of 39 studies that included 4,052 patients, the detection of ctDNA was associated with a significantly worse OS in multivariable analyses [HR 2.70; 95% CI, 2.02–3.61; p < .001).107 There was also a statistically significant association between high total cell-free DNA and worse OS at 3 years by multivariate analysis (HR 1.91; 95% CI, 1.59–2.29; p < .001).107 Thus, the use of liquid biopsy to identify shedders and nonshedders has important clinical implication and has been proposed to be included as part of the staging workup (tumor, node, metastasis, biopsy).10
CONCLUSION
The advantages of liquid biopsy not only include the possibility to circumvent the need to achieve adequate tumor tissue biopsy specimens, it also reflects an aggregate of ctDNA output from potentially both primary and all metastatic sites accounting for tumor heterogeneity that cannot be evaluated by a single core tumor needle biopsy. Liquid biopsy may provide prognostic implication and help guide initiation and/or duration of adjuvant treatment by detecting MRD. Although all the current government-approved liquid biopsy test kits are designed to detect only mutations in one or a few specific genes, the future of liquid biopsy will be using NGS with increased sensitivity and ability to detect all modes of genetic alteration and TMB. The gene panel and limit of detection used by commercial NGS liquid biopsy kits change over time. It is incumbent upon the ordering physician to understand the current NGS platforms and performance characteristics and upon readers to interpret the literature with the comprehension of the changes in the gene panel and performance characteristics of commercial NGS tests as time goes by.
PRACTICAL APPLICATIONS.
Despite the widespread use of liquid biopsy in solid malignancy, only five distinct test kits have government approval: Cobas EGFR Mutations Test v2 (Roche Molecular Diagnostics; FDA approval for detection of EGFR del19, EGFR L858R, and EGFR T790M in lung cancer), Therascreen EGFR RGQ Plasma PCR kit (Qiagen Inc; E.U. approval for detection of EGFR del19 and EGFR L858R in lung cancer); AmoyDx Super-ARMS EGFR mutation test (AmoyDx; Chinese Food and Drug Administration approval for detection of EGFR del19, EGFR 858R, and EGFR T790M in lung cancer); OncoBEAM RAS CRC Kit (Sysmex Inostics; E.U. approval for detection of KRAS and NRAS mutations in colorectal cancer); and Idylla ctKRAS Mutation Test and Idylla ctNRAS-BRAF Mutation Test (Biocartis, Inc.; E.U. approval for detection of KRAS, NRAS, and BRAF mutations in colorectal cancer).
Currently, liquid biopsy is most commonly used to detect actionable EGFR, RAS (KRAS, NRAS), and BRAF V600E mutations, with potentially in the future for detection of PIK3CA mutations.
Liquid biopsy based on NGS can detect multiple genomic alterations (mutations, indel, amplification, rearrangement) in multiple actionable genes and also determine tumor mutation burden used to potentially select for immune checkpoint inhibitors. Eventually, liquid biopsy using NGS will supersede use of single/oligo gene liquid biopsy detection kits.
Liquid biopsy to detect nonspecific ctDNA in tumors without clear driver mutations may be most useful in detecting minimal residual disease, which can account for tumor heterogeneity in the adjuvant setting.
Distinguishing “shedders” and “nonshedders” of ctDNA may provide insight into tumor biology and prognostic significance.
Footnotes
Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.
References
- 1.Wan JCM, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–238. [DOI] [PubMed] [Google Scholar]
- 2.Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Webb S. The cancer bloodhounds. Nat Biotechnol. 2016;34:1090–1094. [DOI] [PubMed] [Google Scholar]
- 4.Newman AM, Lovejoy AF, Klass DM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 2016;34:547–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Oxnard GR, Paweletz CP, Sholl LM. Genomic analysis of plasma cell-free DNA in patients with cancer. JAMA Oncol. 2017;3:740–741. [DOI] [PubMed] [Google Scholar]
- 6.Sacher AG, Komatsubara KM, Oxnard GR. Application of plasma genotyping technologies in non-small cell lung cancer: a practical review. J Thorac Oncol. 2017;12:1344–1356. [DOI] [PubMed] [Google Scholar]
- 7.Huang WL, Chen YL, Yang SC, et al. Liquid biopsy genotyping in lung cancer: ready for clinical utility? Oncotarget. 2017;8:18590–18608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bernabé R, Hickson N, Wallace A, et al. What do we need to make circulating tumour DNA (ctDNA) a routine diagnostic test in lung cancer? Eur J Cancer. 2017;81:66–73. [DOI] [PubMed] [Google Scholar]
- 9.Thierry AR, El Messaoudi S, Mollevi C, et al. Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment. Ann Oncol. 2017;28:2149–2159. [DOI] [PubMed] [Google Scholar]
- 10.Yang M, Forbes ME, Bitting RL, et al. Incorporating blood-based liquid biopsy information into cancer staging: time for a TNMB system? Ann Oncol. 2018;29:311–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.U.S. Food and Drug Administration. Cobas EGFR Mutation Test v2. 2016. https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm504540.htm. Accessed January 15, 2018.
- 12.U.S. Food and Drug Administration. Cobas EGFR mutation test v2. http://www.accessdata.fda.gov/cdrh_docs/pdf12/P120019S007c.pdf. Accessed January 28,2018.
- 13.Malapelle U, Sirera R, Jantus-Lewintre E, et al. Profile of the Roche cobas® EGFR mutation test v2 for non-small cell lung cancer. Expert Rev Mol Diagn. 2017;17:209–215. [DOI] [PubMed] [Google Scholar]
- 14.Mok TS, Wu Y-L, Ahn M-J, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med. 2017;376:629–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wu YL, Zhou C, Liam CK, et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol. 2015;26:1883–1889. [DOI] [PubMed] [Google Scholar]
- 16.Reck M, Hagiwara K, Han B, et al. ctDNA Determination of EGFR mutation status in European and Japanese patients with advanced NSCLC: the ASSESS Study. J Thorac Oncol. 2016;11:1682–1689. [DOI] [PubMed] [Google Scholar]
- 17.Han B, Tjulandin S, Hagiwara K, et al. EGFR mutation prevalence in Asia-Pacific and Russian patients with advanced NSCLC of adenocarcinoma and non-adenocarcinoma histology: the IGNITE study. Lung Cancer. 2017;113:37–44. [DOI] [PubMed] [Google Scholar]
- 18.Jenkins S, Yang JC, Ramalingam SS, et al. Plasma ctDNA analysis for detection of the EGFR T790M mutation in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2017;12:1061–1070. [DOI] [PubMed] [Google Scholar]
- 19.Remon J, Menis J, Hasan B, et al. The APPLE trial: feasibility and activity of AZD9291 (osimertinib) treatment on positive plasma T790M in EGFR-mutant NSCLC patients. EORTC 1613. Clin Lung Cancer. 2017;18:583–588. [DOI] [PubMed] [Google Scholar]
- 20.European Medicines Agency. Iressa: public assessment report — product information. 2016. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/001016/WC500036358.pdf.
- 21.European Medicines Agency. Tagrisso: public assessment report — product information. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR__Product_Information/human/004124/WC500202022.pdf.
- 22.U.S. Food and Drug Administration. Therascreen® EGFR RGQ PCR Kit Instructions for Use (Handbook). https://www.accessdata.fda.gov/cdrh_docs/pdf12/P120022c.pdf. Accessed January 28, 2018.
- 23.Thress KS, Brant R, Carr TH, et al. EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer. 2015;90:509–515. [DOI] [PubMed] [Google Scholar]
- 24.Douillard JY, Ostoros G, Cobo M, et al. First-line gefitinib in Caucasian EGFR mutation- positive NSCLC patients: a phase-IV, open-label, single-arm study. Br J Cancer. 2014;110:55–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Douillard JY, Ostoros G, Cobo M, et al. Gefitinib treatment in EGFR mutated Caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol. 2014;9:1345–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.AmoyDx. AmoyDx Super-BEAM EGFR detection kit. http://www.amoydiagnostics.com/China-FDA-Grants-Liquid-Biopsy-Approval-to-AmoyDx-Super-ARMS-EGFR-Mutation-Test_n114. Accessed February 1, 2018.
- 27.Li Y, Xu H, Su S, et al. Clinical validation of a highly sensitive assay to detect EGFR mutations in plasma cell-free DNA from patients with advanced lung adenocarcinoma. PLoS One. 2017;12:e0183331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhou C, Wang M, Cheng Y, et al. Detection of EGFR T790M in Asia-Pacific patients (pts) with EGFR mutation-positive advanced non-small cell lung cancer (NSCLC): circulating tumour (ctDNA) across 3 platforms. Ann Oncol. 2017;28(suppl_5):v460–v496. [Google Scholar]
- 29.Yang JC, Sequist LV, Geater SL, et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol. 2015;16: 830–838. [DOI] [PubMed] [Google Scholar]
- 30.National Institute for Health and Care Excellence. Plasma EGFR mutation tests for adults with locally advanced or metastatic non-small-cell lung cancer. Medtech Innovation Briefing. January 18, 2018. https://www.nice.org.uk/advice/mib137/resources/plasma-egfr-mutation-tests-for-adults-with-locally-advanced-or-metastaticnonsmallcell-lung-cancer-pdf-2285963400737221. Accessed January 28, 2018.
- 31.Gray JE, Okamoto I, Sriurapong V, et al. Osimertinib vs SoC EGFR-TKI as first-line treatment in patients with EGFRm advanced NSCLC (FLAURA): plasma ctDNA analysis. J Thorac Oncol. 2017;12:S1754–S1755. [DOI] [PubMed] [Google Scholar]
- 32.Lin CC, Shih JY, Yu CJ, et al. Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: a genomic study. Lancet Respir Med. 2018;6:107–116. [DOI] [PubMed] [Google Scholar]
- 33.Karachaliou N, Mayo-de las Casas C, Queralt C, et al. Association of EGFR L858R mutation in circulating free DNA with survival in the EURTAC trial. JAMA Oncol. 2015;1:149–157. [DOI] [PubMed] [Google Scholar]
- 34.Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239–246. [DOI] [PubMed] [Google Scholar]
- 35.Douillard JY, Oliner KS, Siena S, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–1034. [DOI] [PubMed] [Google Scholar]
- 36.Van Cutsem E, Lenz HJ, Köhne CH, et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 2015;33:692–700. [DOI] [PubMed] [Google Scholar]
- 37.Sysmex Inostics. OncoBEAMTM RAS CRC Kit Instructions for Use. https://www.sysmex-inostics.com/products/product-single-view/oncobeamTM-ras-crc-kit-ivd-3855.html. Accessed February 1, 2018.
- 38.Grasselli J, Elez E, Caratu G, et al. Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol. 2017;28:1294–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vidal J, Muinelo L, Dalmases A, et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol. 2017;28:1325–1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Schmiegel W, Scott RJ, Dooley S, et al. Blood-based detection of RAS mutations to guide anti-EGFR therapy in colorectal cancer patients: concordance of results from circulating tumor DNA and tissue-based RAS testing. Mol Oncol. 2017;11:208–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Normanno N, Abate RE, Lambiase M, et al. RAS testing of liquid biopsy correlates with the outcome of metastatic colorectal cancer patients treated with first-line FOLFIRI plus cetuximab in the CAPRI-GOIM trial. Ann Oncol. 2018;29:112–118. [DOI] [PubMed] [Google Scholar]
- 42.Hahn S, Tannapfel A, Friess H, et al. Clinical performance comparison of RAS mutation detection in plasma and tissue samples from stage IV colorectal cancer patients from first-line anti-EGFR therapy selection. Eur J Cancer. 2015;51:S79. [Google Scholar]
- 43.Ma W. Significant improvement in detecting BRAF, KRAS, and EGFR mutations using next-generation sequencing as compared with FDA-cleared kits. Mol Diagn Ther. 2017;21:571–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Garcia-Foncillas J, Alba E, Aranda E, et al. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: an expert taskforce review. Ann Oncol. 2017;28:2943–2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Jacobs B, Claes B, Bachet JB, et al. Evaluation of a fully automated extended RAS-BRAF test on prospectively collected plasma samples from patients with metastatic colorectal cancer. J Clin Oncol. 2017;35(15_suppl):e15127. [Google Scholar]
- 46.Bachet JB, Bouche O, Taïeb J, et al. RAS mutations concordance in circulating tumor DNA (ctDNA) and tissue in metastatic colorectal cancer (mCRC): RASANC, an AGEO prospective multicenter study. J Clin Oncol. 2017;35(15_suppl):11509. [Google Scholar]
- 47.Braig F, Voigtlaender M, Schieferdecker A, et al. Liquid biopsy monitoring uncovers acquired RAS-mediated resistance to cetuximab in a substantial proportion of patients with head and neck squamous cell carcinoma. Oncotarget. 2016;7:42988–42995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Van Emburgh BO, Arena S, Siravegna G, et al. Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat Commun. 2016;7:13665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pietrantonio F, Vernieri C, Siravegna G, et al. Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. Clin Cancer Res. 2017;23:2414–2422. [DOI] [PubMed] [Google Scholar]
- 51.Kim G, McKee AE, Ning YM, et al. FDA approval summary: vemurafenib for treatment of unresectable or metastatic melanoma with the BRAFV600E mutation. Clin Cancer Res. 2014;20:4994–5000. [DOI] [PubMed] [Google Scholar]
- 52.Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. 2017;18:1307–1316. [DOI] [PubMed] [Google Scholar]
- 53.Gonzalez-Cao M, Mayo-de-Las-Casas C, Molina-Vila MA, et al. BRAF mutation analysis in circulating free tumor DNA of melanoma patients treated with BRAF inhibitors. Melanoma Res. 2015;25:486–495. [DOI] [PubMed] [Google Scholar]
- 54.El Messaoudi S, Mouliere F, Du Manoir S, et al. Circulating DNA as a strong multimarker prognostic tool for metastatic colorectal cancer patient management care. Clin Cancer Res. 2016;22:3067–3077. [DOI] [PubMed] [Google Scholar]
- 55.Baselga J, Im SA, Iwata H, et al. Buparlisib plus fulvestrant versus placebo plus fulvestrant in postmenopausal, hormone receptor-positive, HER2-negative, advanced breast cancer (BELLE-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18:904–916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Di Leo A, Johnston S, Lee KS, et al. Buparlisib plus fulvestrant in postmenopausal women with hormone-receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018;19:87–100. [DOI] [PubMed] [Google Scholar]
- 57.Chung TKH, Cheung TH, Yim SF, et al. Liquid biopsy of PIK3CA mutations in cervical cancer in Hong Kong Chinese women. Gynecol Oncol. 2017;146:334–339. [DOI] [PubMed] [Google Scholar]
- 58.Christensen E, Birkenkamp-Demtröder K, Nordentoft I, et al. Liquid biopsy analysis of FGFR3 and PIK3CA hotspot mutations for disease surveillance in bladder cancer. Eur Urol. 2017;71:961–969. [DOI] [PubMed] [Google Scholar]
- 59.Kidess-Sigal E, Liu HE, Triboulet MM, et al. Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget. 2016;7:85349–85364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. [DOI] [PubMed] [Google Scholar]
- 61.Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–697. [DOI] [PubMed] [Google Scholar]
- 62.Shoda K, Ichikawa D, Fujita Y, et al. Monitoring the HER2 copy number status in circulating tumor DNA by droplet digital PCR in patients with gastric cancer. Gastric Cancer. 2017;20:126–135. [DOI] [PubMed] [Google Scholar]
- 63.Shoda K, Masuda K, Ichikawa D, et al. HER2 amplification detected in the circulating DNA of patients with gastric cancer: a retrospective pilot study. Gastric Cancer. 2015;18:698–710. [DOI] [PubMed] [Google Scholar]
- 64.Kim ST, Banks KC, Lee SH, et al. Prospective feasibility study for using cell-free circulating tumor DNA–guided therapy in refractory metastatic solid cancers: an interim analysis. Precision Oncol. 2017;1:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fribbens C, O’Leary B, Kilburn L, et al. Plasma ESR1 mutations and the treatment of estrogen receptor-positive advanced breast cancer. J Clin Oncol. 2016;34:2961–2968. [DOI] [PubMed] [Google Scholar]
- 66.Fribbens C, Garcia Murillas I, Beaney M, et al. Tracking evolution of aromatase inhibitor resistance with circulating tumour DNA analysis in metastatic breast cancer. Ann Oncol. 2018;29:145–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chung JH, Pavlick D, Hartmaier R, et al. Hybrid capture-based genomic profiling of circulating tumor DNA from patients with estrogen receptor-positive metastatic breast cancer. Ann Oncol. 2017;28:2866–2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Antonarakis ES, Lu C, Wang H, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Azad AA, Volik SV, Wyatt AW, et al. Androgen receptor gene aberrations in circulating cell-free NA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin Cancer Res. 2015;21:2315–2324. [DOI] [PubMed] [Google Scholar]
- 70.Wyatt AW, Azad AA, Volik SV, et al. Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer. JAMA Oncol. 2016;2:1598–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Guardant Health. Guardant 360 http://www.guardanthealth.com. Accessed December 31, 2017
- 72.Foundation Medicine. Genomic Testing. FoundationACT. https://www.foundationmedicine.com/genomic-testing/foundation-act. Accessed December 31, 2017.
- 73.Lanman RB, Mortimer SA, Zill OA, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS One. 2015;10:e0140712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kim ST, Lee WS, Lanman RB, et al. Prospective blinded study of somatic mutation detection in cell-free DNA utilizing a targeted 54-gene next generation sequencing panel in metastatic solid tumor patients. Oncotarget. 2015;6:40360–40369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kato S, Kurasaki K, Ikeda S, Kurzrock R. Rare tumor clinic: the University of California San Diego Moores Cancer Center experience with a precision therapy approach. Oncologist. 2018;23:171–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kato S, Krishnamurthy N, Banks KC, et al. Utility of genomic analysis in circulating tumor DNA from patients with carcinoma of unknown primary. Cancer Res. 2017;77:4238–4246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hahn AW, Gill DM, Maughan B, et al. Correlation of genomic alterations assessed by next-generation sequencing (NGS) of tumor tissue DNA and circulating tumor DNA (ctDNA) in metastatic renal cell carcinoma (mRCC): potential clinical implications. Oncotarget. 2017;8:33614–33620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Riviere P, Fanta PT, Ikeda S, et al. The mutational landscape of gastrointestinal malignancies as reflected by circulating tumor DNA. Mol Cancer Ther. 2018;17:297–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Blakely CM, Watkins TBK, Wu W, et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet. 2017;49:1693–1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kuderer NM, Burton KA, Blau S, et al. Comparison of 2 commercially available next-generation sequencing platforms in oncology. JAMA Oncol. 2017;3:996–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Schwaederle M, Chattopadhyay R, Kato S, et al. Genomic alterations in circulating tumor DNA from diverse cancer patients identified by next-generation sequencing. Cancer Res. 2017;77:5419–5427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Torga G, Pienta KJ. Patient-paired sample congruence between 2 commercial liquid biopsy tests. JAMA Oncol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Maia MC, Bergerot PG, Dizman N, et al. Association of circulating tumor DNA (ctDNA) detection in metastatic renal cell carcinoma (mRCC) with tumor burden. Kidney Cancer. 2017;1:65–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ou SI, Lee TK, Young L, et al. Dual occurrence of ALK G1202R solvent front mutation and small cell lung cancer transformation as resistance mechanisms to second generation ALK inhibitors without prior exposure to crizotinib. Pitfall of solely relying on liquid re-biopsy? Lung Cancer. 2017;106:110–114. [DOI] [PubMed] [Google Scholar]
- 85.Ou SI, Cui J, Schrock AB, et al. Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796 (G796S/R) together with C797S/R and L792F/H mutations in one EGFR (L858R/T790M) NSCLC patient who progressed on osimertinib. Lung Cancer. 2017; 108:228–231. [DOI] [PubMed] [Google Scholar]
- 86.Ou SI, Horn L, Cruz M, et al. Emergence of FGFR3-TACC3 fusions as a potential by-pass resistance mechanism to EGFR tyrosine kinase inhibitors in EGFR mutated NSCLC patients. Lung Cancer. 2017;111:61–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Klempner SJ, Mehta P, Schrock AB, et al. Cis-oriented solvent-front EGFR G796S mutation in tissue and ctDNA in a patient progressing on osimertinib: a case report and review of the literature. Lung Cancer (Auckl). 2017;8:241–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.ThermoFisher Scientific. Oncomine Lung cfDNA assay. https://www.thermofisher.com/order/catalog/product/A31149. Accessed December 31, 2017.
- 89.Archer Reveal. ctDNA 28 Liquid biopsy mutation profiling with Archer Reveal ctDNA 28 http://archerdx.com/reveal-ctdna. Accessed December 31, 2017.
- 90.Genomic Health. Oncotype SEQ Liquid Select. http://www.genomichealth.com/en-US/oncotype_iq_products/oncotype_SEQ. Accessed December 31, 2017.
- 91.MolecularMD. Platforms, Applications & Methods. http://molecularmd.com/platforms-applications-methods/overview/. Accessed December 31, 2017. [Google Scholar]
- 92.Pathway Genomics. Pathway Genomics launches first liquid biopsy test to detect cancer-associated mutations in high-risk patients. https://www.pathway.com/pathway-genomics-launches-first-liquid-biopsy-test-to-detect-cancer-associated-mutations-in-high-risk-patients/. Accessed December 31, 2017.
- 93.DiCarta. OptiSeq NGS Pan-cancer Panel (65 Genes). http://www.diacarta.com/services/ngs/. Accessed December 12, 2018
- 94.Personal Genome Diagnostics. MetDETECT R. http://www.personal-genome.com/pharma-biotech-companies/liquid-biopsy/. Accessed December 12, 2018.
- 95.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;9:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Koeppel F, Blanchard S, Jovelet C, et al. Whole exome sequencing for determination of tumor mutation load in liquid biopsy from advanced cancer patients. PLoS One. 2017;12:e0188174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fabrizio D, Malboeuf C, Lieber D, et al. Analytic validation of a next generation sequencing assay to identify tumor mutational burden from blood (bTMB) to support investigation of an anti-PD-L1 agent, atezolizumab, in a first line non-small cell lung cancer trial (BFAST). Ann Oncol. 2017;28(suppl_5):v22–v42. [Google Scholar]
- 99.Gandara DR, Kowanetz M, Mok TS, et al. Blood-based biomarkers for cancer immunotherapy: tumor mutational burden in blood (bTMB) is associated with improved atezolizumab (atezo) efficacy in 2L+ NSCLC (POPLAR and OAK). Ann Oncol. 2017; 28(suppl_5):mdx380. [Google Scholar]
- 100.Mok TS, Gadgeel S, Kim ES, et al. 1383TiP Blood first line ready screening trial (B-F1RST) and blood first assay screening trial (BFAST) enable clinical development of novel blood-based biomarker assays for tumor mutational burden (TMB) and somatic mutations in 1L advanced or metastatic NSCLC. Ann Oncol. 2017; 28(suppl_5):mdx380.084. [Google Scholar]
- 101.Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med. 2016;8: 346ra92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature. 2017;545:446–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the evolution of non-small-cell lung cancer. N Engl J Med. 2017;376:2109–2121. [DOI] [PubMed] [Google Scholar]
- 104.Murphy DJ, Blyth KG. Predicting lung cancer recurrence from circulating tumour DNA. Commentary on ‘Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution’. Cell Death Differ. 2017;24:1473–1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ng TL, Camidge DR. Lung cancer’s real adjuvant EGFR targeted therapy questions. Lancet Oncol. 2018;19:15–17. [DOI] [PubMed] [Google Scholar]
- 106.Chan AT, Lo YM, Zee B, et al. Plasma Epstein-Barr virus DNA and residual disease after radiotherapy for undifferentiated nasopharyngeal carcinoma. J Natl Cancer Inst. 2002;94:1614–1619. [DOI] [PubMed] [Google Scholar]
- 107.Ocana A, Diez-Gonzalez L, Garcia-Olmo DC, et al. Circulating DNA and survival in solid tumors. Cancer Epidemiol Biomarkers Prev. 2016;25:399–406. [DOI] [PubMed] [Google Scholar]