Abstract
Valeriy Domenyuk1,*, Kasey Benson1,*, Peggy Carter1, Daniel Magee1, Jian Zhang1, Nitin Bhardwaj1, Hongseok Tae1, James Wacker1, Foram Rathi1, Siobhan Miick1, Aastha Kohli1, Joshua Carroll1, Lori Cuyugan1, Evelyn Perez1, Wayeesha Zhang1, John Collins1, Patrick Kennedy1, Jeremy Ellis1, Adam Stark1, Andrey Loskutov1, Brittany Cuttone1, Blake Taylor1, Rebecca Feldman1, Jeff Swenson1, David Bryant1, Robert Hahn-Lowry1, Raunaq Kaushal1, Jennifer R. Ribeiro1, Jim Abraham1, Milan Radovich1, George W. Sledge1, Matthew Oberley1 and David Spetzler1
1 Caris Life Sciences, Irving, TX 75039, USA
* These authors contributed equally to this work
Correspondence to:
David Spetzler, | email: | [email protected] |
Keywords: companion diagnostic; molecular profiling; solid tumors; precision oncology; next-generation sequencing
Received: April 10, 2025 Accepted: July 30, 2025 Published: August 13, 2025
ABSTRACT
The precision oncology industry has evolved rapidly within the past two decades, although treatment selection remains a complex undertaking. Access to timely, accurate, and comprehensive molecular profiling data is imperative to improving patient outcomes within the expanding sphere of Food and Drug Administration (FDA)-approved targeted therapies. Caris Life Sciences has developed MI Cancer Seek®, an FDA-approved whole exome and whole transcriptome sequencing-based molecular test encompassing adult and pediatric tumor profiling, eight companion diagnostics (CDx), and additional laboratory developed test (LDT) capabilities. Patient tissue is maximized through simultaneous analysis of DNA and RNA with minimum input of 50 ng. The clinical and analytical validation presented herein demonstrates non-inferiority of MI Cancer Seek relative to other FDA-approved CDx tests (>97% negative and positive percent agreement), as well as its precision, sensitivity, and specificity. Accordingly, MI Cancer Seek represents a safe and effective comprehensive molecular test option supporting biomarker-directed care for oncology patients.
Introduction
Precision oncology aims to improve patient outcomes through coupling tumor molecular profiling to specific targeted therapies. Although the number of United States Food and Drug Administration (FDA)-approved targeted therapies available to patients continues to rapidly expand, decisions regarding the use of specific therapies for select patients can be complex. Significant effort has been given to the development and application of genomic testing assays to decipher a heterogeneous molecular landscape of cancer, thus assisting clinicians in making informed therapeutic choices for patients [1–7]. The growing clinical utility of comprehensive multi-gene panels for oncology treatment planning is reflected in current recommendations by The American Society of Clinical Oncology (ASCO) [8], The European Society for Medical Oncology (ESMO) [9], and the National Comprehensive Cancer Network (NCCN) [10–14].
Sequencing of tumor DNA and RNA creates a complex portrait of the tumor genome, including identification of pathogenic variants and tumor signatures [15]. Next-generation sequencing (NGS) technologies, such as whole exome sequencing (WES), have been increasingly available in the clinical setting, while improved tumor excision and sample processing techniques have allowed for better characterization of even degraded nucleic acid found in formalin-fixed paraffin embedded (FFPE) tissues [16–18]. Whole transcriptome sequencing (WTS) can provide additional information about specific molecular alterations as well as gene expression, adding another layer of information that clinicians can use to identify appropriate treatments or clinical trials. However, techniques analyzing DNA and RNA have traditionally been performed independently [19], requiring sufficient quantities of tumor tissue to satisfy quality standards and input requirements for molecular assays.
Here, we present the clinical and analytical validation of the Molecular Intelligence (MI) Cancer Seek® assay, developed at Caris Life Sciences, a College of American Pathologists (CAP)-accredited and Clinical Laboratory Improvement Amendments (CLIA)-certified laboratory. This FDA-approved (P240010) comprehensive molecular profiling assay is the evolution of the laboratory developed test (LDT) MI Tumor Seek Hybrid®, which has been in clinical use since February 2023. MI Cancer Seek analyzes tumor DNA and RNA simultaneously from a single total nucleic acid (TNA) extraction and targets eight companion diagnostic (CDx) indications: PIK3CA alterations in breast cancer; KRAS/NRAS wild-type status in colorectal carcinoma (CRC); BRAFV600E mutations in CRC; BRAFV600E or BRAFV600K mutations in melanoma; EGFR exon19 deletions and L858R mutations in NSCLC; and microsatellite instability (MSI) status in endometrial carcinoma (not MSI-high; i.e., microsatellite stable (MSS)) and solid tumors (MSI-high) [20]. These indications are associated with FDA-approved targeted therapies (Table 1), based on pivotal clinical trial evidence [21–27].
Table 1: MI Cancer Seek CDx indications
Tumor type | Biomarker(s) and specific alteration(s) detected | Therapy |
---|---|---|
Breast (DNA only for PIK3CA) | PIK3CA (C420R; E542K; E545A, E545D [1635G>T only], E545G, E545K, Q546E, Q546R; and H1047L, H1047R, H1047Y) | PIQRAY® (alpelisib) |
Colorectal Cancer (CRC) (DNA only) | KRAS wild-type (absence of mutations in exons 2, 3, and 4) and NRAS wild-type (absence of mutations in exons 2, 3, and 4) | VECTIBIX® (panitumumab) |
BRAF V600E | BRAFTOVI® (encorafenib) in combination with ERBITUX® (cetuximab) | |
Melanoma (DNA only) | BRAF V600E | BRAF Inhibitors approved by FDAa |
BRAF V600E or V600K | MEKINIST® (trametinib) or BRAF/MEK Inhibitor Combinations approved by FDAa | |
Non-small Cell Lung Cancer (NSCLC) (DNA only) | EGFR activating mutations: EGFR exon 19 deletions and exon 21 (L858R) alterations | EGFR Tyrosine Kinase Inhibitors approved by FDAa |
Solid Tumors | MSI-high | KEYTRUDA® (pembrolizumab), JEMPERLI® (dostarlimab-gxly) |
Endometrial Carcinoma | Microsatellite stable (MSS) (Not MSI-high) | KEYTRUDA® (pembrolizumab) in combination with LENVIMA® (lenvatinib) |
The ‘hybrid’ methodology of MI Cancer Seek allows efficient tissue utilization and accurate and comprehensive detection of a wide range of clinically relevant biomarkers from a minimum DNA input of 50 ng isolated from microdissected, FFPE tumor tissue with minimum 20% tumor content (Figure 1). DNA is sequenced to an average depth of 230× for 20,859 genes (whole exome), 1000× for 720 genes with known and potential clinical relevance, 1500× for 228 reportable genes, and RNA to a minimum of 1.37 million total mapped reads. An annotated clinical report is generated including biomarkers that have a high-level of evidence to associate with matched FDA-approved therapies, as well as additional clinically relevant information based on published evidence. The validation presented here demonstrates that MI Cancer Seek is a robust, accurate, and precise molecular test that meets FDA standards for CDx indications and tumor mutational profiling, with additional LDT capabilities as described.
Figure 1: Overview of MI Cancer Seek workflow.
MI Cancer Seek begins with total nucleic acid (TNA) extraction from formalin-fixed, paraffin-embedded (FFPE) tissue slides. A minimum 20% tumor content and 25 mm2 is required, which is obtained through manual microdissection if required. During library preparation, RNA is labeled during first strand cDNA synthesis. Sequencing is performed on qualified Illumina NovaSeq 6000 instruments. Raw data is processed by Caris’ proprietary bioinformatics pipeline, and a report is generated that includes CDx biomarkers (level 1 evidence), clinically relevant biomarkers (level 2 evidence), and biomarkers with possible clinical significance (level 3 evidence). Abbreviations: CNV: copy number variation; EBV: Epstein-Barr virus; GPS: genomic probability score; HLA: human leukocyte antigen; HPV: human papilloma virus; HRD: homologous recombination deficiency; INDEL: insertion/deletion; LoH: loss of heterozygosity; MCPyV: Merkel cell polyomavirus; MSI: microsatellite instability; SNV: single nucleotide variant; TMB: tumor mutational burden. Created in BioRender. Ribeiro, J. (2025) https://BioRender.com/m29z318.
Results
Clinical validation for FDA companion diagnostic (CDx) and tumor profiling claims
Non-inferiority studies for CDx claims
MI Cancer Seek has eight CDx claims representing high clinical burden in the oncology patient population (Table 1, Supplementary Table 1). As a follow-on CDx (FCD) device, clinical validity was demonstrated through non-inferiority to an appropriate FDA-approved test (Supplementary Table 2), as described by Li et al. [28]. One replicate tested by MI Cancer Seek (FCD replicate) was compared to two replicates tested by each comparator companion diagnostic (CCD) test for each CDx claim, as shown in Figure 2. The results for FCD comparison to concordant CCD replicates are summarized in Table 2, and full concordance results are shown in Supplementary Tables 3–5. Regarding PIK3CA alterations in breast cancer, KRAS/NRAS wild-type status in advanced CRC, BRAFV600E mutations in CRC, BRAFV600E or BRAFV600K mutations in melanoma, EGFR exon19 deletions and L858R mutations in NSCLC, and MSI status in endometrial carcinoma and solid tumors, positive percent agreement (PPA) and negative percent agreement (NPA) ranged from 97–100%, and all upper limits of the 95% confidence intervals met pre-specified non-inferiority margins, as described in ‘Methods’.
Figure 2: Schematic of MI Cancer Seek validation for CDx claims.
Remnant samples were enrolled meeting study criteria. Samples were de-identified and lab personnel blinded to their biomarker status. Unstained slides (USS) were used to complete two tests with the comparator companion diagnostic (CCD) and one test with MI Cancer Seek as the follow-on companion diagnostic (FCD). Concordance was evaluated by determining the agreement between the two CCD replicates to the agreements of FCD to CCD1 and FCD to CCD2. Concordance with FCD was also determined using the concordance between both CCD replicates as “truth”. Non-inferiority was the primary endpoint for analysis.
Table 2: Summary of PPA and NPA for MI Cancer Seek CDx clinical concordance studies
Biomarker and indication | FDA-approved Comparator Method | Sample Size, Na | PPA (95% CI) | NPA (95% CI) |
---|---|---|---|---|
MSI Status in Solid Tumor Types | Ventana MMR RxDx Panel | 401 | 97.5% (94, 99.1) | 98.5% (95.4, 99.7) |
MSI Status in EC | Ventana MMR RxDx Panel | 251 | 98.4% (93.8, 99.9) | 97.6% (92.8, 99.5) |
BRAFV600E/K in Melanoma | bioMérieux THxID BRAF Kit | 330 | 98.9% (95.6, 99.9) | 99.3% (95.9, 100.2) |
BRAFV600E in CRC | therascreen BRAF V600E RGQ PCR Kit | 352 | 99.4% (96.5, 100.2) | 100% (97.3, 100.4) |
KRAS and NRAS wild-type in CRC | Praxis Extended RAS Panel | 262 | 100.0% (96.1, 100.6) | 97.2% (92.7, 99.1) |
EGFR exon 19 deletions or L858R mutations in NSCLC | Roche cobas EGFR Mutation Test V2 | 315 | 98.1% (91.4, 99.6) | 99.4% (96.1, 100.2) |
PIK3CA alterations in BC | therascreen PIK3CA RGQ PCR Kit | 343 | 99.4% (96.4, 100.2) | 100.0% (97.2, 100.4) |
Tumor profiling validation
MI Cancer Seek provides tumor profiling for use by qualified health professionals in accordance with professional oncology guidelines. Reportable genes with boosted bait coverage are listed in Supplementary Table 6. In order to validate the detection of single nucleotide variants (SNVs) and insertions/deletions (INDELs), MI Cancer Seek results were compared to PGDx elio™ tissue complete, an FDA-cleared targeted gene panel assay [29, 30]. Among 454 eligible specimens, PPA was ≥95% and NPA was 100% for all variant types (Figure 3A, 3B). These metrics demonstrated strong concordance of MI Cancer Seek with the comparator assay. Where discordance was noted, this was determined primarily to be due to inherent differences between the two assays with regards to percent variant frequency (VF) cutoff and variant calling rules, and not due to true discordance between MI Cancer Seek and PGDx elio tissue complete. The ability of MI Cancer Seek to detect ERBB2 (HER2) copy number amplifications (CNAs) in breast cancer was also analyzed. MI Cancer Seek results for ERBB2 CNAs were compared to results of the PathVysion HER2 DNA Probe Kit, yielding 84.7% PPA and 99.4% NPA (Figure 3B). Due to the lower PPA for this comparison, additional clinical investigation to confirm the presence of ERBB2 CNAs with another FDA-approved or cleared test is strongly recommended.
Figure 3: Tumor profiling validations.
(A) Validation of variant detection by MI Cancer Seek compared to PGDx elio tissue complete and ERBB2 copy number amplification compared to fluorescent in situ hybridization (FISH). Numbers of true positive (TP), true negative (TN), false positive (FP), and false negative (FN) are shown. One intermediate sample was counted as positive for ERBB2. One sample that was invalid by MCS and five samples that were invalid by FISH were excluded for ERBB2. (B) Positive percent agreement (PPA) and negative percent agreement (NPA) between MI Cancer Seek and the comparator assay are shown for each variant type. (C) Deming regression analysis of MI Cancer Seek (MCS) TMB values versus the orthogonal test values. Abbreviations: INDEL: insertion/deletion; MNV: multi nucleotide variant; SNV: single nucleotide variant; TMB: tumor mutational burden.
To validate MI Cancer Seek detection of tumor mutational burden (TMB), results from 497 eligible specimens were compared to those from a WES assay (Personalis, Inc., Menlo Park, CA, USA). Deming regression analysis demonstrated a strong correlation between MI Cancer Seek and the Personalis, Inc. WES assay (Pearson r = 0.990; slope = 0.856; intercept = −0.859) (Figure 3C). When comparing TMB using only a subset of exome regions where both assays had ≥50× depth, the concordance between MCS and the orthogonal assay improved even further (slope = 0.927; intercept = −0.68).
Analytical sensitivity of MI Cancer Seek
Analytical sensitivity of MI Cancer Seek was evaluated through limit of detection (LoD) studies to identify the lowest VF (SNVs, INDELs) and lowest tumor content at which at least 95% of the test replicates produced a positive result. CDx biomarkers (including BRAF, EGFR, PIK3CA, and RAS mutations) and other tumor profiling biomarkers (including clinically relevant alterations and TMB) were represented in marker positive samples diluted with lineage matched marker negative samples. Observed VF was concordant with the target VF for each biomarker and reproducible across replicates (Supplementary Figure 1). The results of LoD studies support the recommended sensitivity of variant detection at >5% VF for MI Cancer Seek, illustrated by reduced detection rates at 5% VF for certain mutations (Figure 4A). LoD for ERBB2 CNAs was determined to be 8.3 copies, which was the lowest level of copies yielding ≥95% detection (Figure 4B). Furthermore, tumor content LoD studies for MSI and TMB found reliable detection of these biomarkers at 20% cutoff (Figure 4C, 4D), which exceeded the tumor content LoD for other alterations (15% for ERBB2 CNAs, 10.5% for all SNVs and INDELs, and 12.0% for CDx variants) (Supplementary Table 7). Thus, the 20% tumor content requirement for MI Cancer Seek is considered sufficient to enable reliable detection of all variants/biomarkers.
Figure 4: Limit of detection (LoD).
(A) LoD for variant frequency (VF) of SNVs and INDELs (including CDx biomarkers). Caris cutoff is 5% VF for reporting. (B) Copies LoD for ERBB2 CNA. (C, D) LoD for % tumor content (TC) including MSI (C), and TMB (D). Hit rate (%) at each given VF (A), nReads (B) or tumor content (C) is shown by the heatmap color, with the quantitative hit rate (%) shown in each box. The coefficient of variation (% CV) is shown for TMB in (D). The red boxes highlight the LoD cutoff employed by MI Cancer Seek. In (C, D), the quantitative values for MSI (C) and TMB (D) are indicated in parentheses. Abbreviations: BC: breast carcinoma; CNA: copy number amplification; CRC: colorectal carcinoma; EC: endometrial carcinoma; INDEL: insertion/deletion; Mel: melanoma; MSI: microsatellite instability; NSCLC: non-small cell lung cancer; OSEC: ovarian surface epithelial carcinoma; PC: prostate carcinoma; TC: tumor content; SNV: single nucleotide variant; VF: variant frequency.
A limit of blank (LoB) study established the false positive rate using non-tumor wild-type (WT) samples across multiple tissue types. The results demonstrate a false positive rate of 0% for most DNA capabilities (INDELs, MSI, TMB) and <0.01% for SNVs (Supplementary Table 8). Together, there were a total of three false positive calls for TERTc.-124C>T, which were found to have VF near the variant calling threshold for MI Cancer Seek.
Finally, we performed a DNA input study to investigate the performance of MI Cancer Seek at minimum (50 ng) and optimal/maximum (220 ng) DNA input, as well as suboptimal and overloading levels (25 ng, 37.5 ng, 275 ng, and 330 ng). The PPA and NPA for SNVs, INDELs, MSI, and ERBB2 CNAs were 100% between minimum and optimal DNA input (Table 3). At other input levels, PPA and NPA were also 100% aside from one false negative call for MSI at 37.5 ng. In addition, mean TMB was consistent between 50 ng and 220 ng: the upper bound of the 95% confidence interval (CI) for absolute percent difference in mean TMB values between control and challenge input ranged from 0.0–10.5% (Table 3 and Supplementary Figure 2).
Table 3: DNA input study (Concordance of 50 ng with 220 ng)
Alteration | PPA (95% CI) | NPA (95% CI) | Absolute % Difference in upper bound of CI |
---|---|---|---|
MSI | 100 % (66.4, 100) | 100 % (92.1, 100) | N/A |
INDEL | 100 % (73.5, 100) | 100 % (100, 100) | N/A |
SNV | 100 % (85.8, 100) | 100 % (100, 100) | N/A |
ERBB2 CNA | 100% (73.5, 100) | 100% (92.1, 100) | N/A |
TMB | N/A | N/A | 0–10.5% |
Analytical specificity of MI Cancer Seek
We next evaluated analytical specificity through several studies including interfering substances (exogenous and endogenous), carry-over, and cross-contamination. Exogenous substances included paraffin, xylene, Proteinase K, and 80% ethanol, spiked at the appropriate step to mimic carryover of the substance being tested. Endogenous substances included hemoglobin, colloid, calcium/calcium phosphate, mucin, and conjugated bile acids. The impact of necrotic tissue, melanin, and fat cells on assay performance will be evaluated in a post-market study, although some of these are mitigated whenever possible through microdissection of tumor tissue. There was minimal or no effect detected for exogenous or endogenous substances on assay performance in intended tumor types (Supplementary Table 9). Likewise, carry-over (run-to-run) and cross-contamination (within run) were evaluated at the highest DNA input of 220 ng, which demonstrated 100% PPA and NPA for global mutations, SNVs, INDELs, and MSI. Further specificity studies showed that for any sample, there was a <0.004% chance of index cross-contamination (misassignment of reads to a sample due to both sample indexes being incorrectly switched to another sample). An in silico analysis demonstrated low off-target depth of coverage (mean 1.5×), which confirmed capture bait specificity for reportable gene regions.
Precision of MI Cancer Seek
Next, we performed a precision analysis at low (1–1.5×) and high (2–3×) LoD levels to evaluate the total within-lab variability expected in the MI Cancer Seek assay by evaluating both the individual and combined impact of reagent lots, instruments, operators, and non-consecutive run days across a panel of samples containing representative types of genetic alterations detected by the test. For SNVs, multi-nucleotide variants (MNVs), and INDELs, precision increased according to variant buckets (≥5%, ≥8%, ≥10%, and ≥15% VF), but exceeded 96% even at the lowest VF bucket (Figure 5A). Sample level precision analysis also revealed overall high positive and negative call rates at high and low LoD, although some samples had reduced positive call rates at low LoD (Figure 5B). All CDx markers achieved 100% agreement across replicates except for three cases that had borderline MSI status resulting in agreements of 97.2%, 91.7%, and 94.4%. Coefficient of variation (CV), which is the ratio of the standard deviation to the mean and is a measure of assay precision, was determined for MSS/MSI-high (measured by quantitative loci) and TMB (measured across a range of values from 2.3–30.2), incorporating operator/instrument, reagent lot, within-run, and between-run variability (Figure 5C–5F). Together, the results of these analyses support the precision of MI Cancer Seek for tumor profiling and CDx biomarker detection at LoD thresholds.
Figure 5: Precision study.
There were 49 panel members in the precision study. Each panel member was tested a total of 36 times, including three operator teams, three instrument sets, three reagent lots, and was minimally executed across three non-consecutive days over a 20-day span. (A) Precision per variant frequency (VF) bucket for INDELs, MNVs, and SNVs. Variants included passenger variants that were not the basis of panel member selection. (B) Sample-level positive and negative calls rates for tracked variants in all 49 panel members in precision study. There were 1–19 variants per sample. 37 panel members were tested at high (2–3×) and low (1–1.5×) LoD. One additional panel member was tested only at high LoD and one additional panel member only at low LoD. The remaining panel members did not have variant frequency LoD adjusted (two panel members were MSS, four panel members were MSI-high, two panel members were TMB-high, and two panel members had an ERBB2 CNA). (C) Mean MSI loci (n = 6) is shown with total within-lab standard deviation, including operator/instrument, reagent lot, within-run, and between-run variability. (D) %CV for MSI loci is shown. (E) Mean TMB (n = 49) is shown with total within-lab standard deviation, including operator/instrument, reagent lot, within-run, and between-run variability. TMB was measured in all 86 samples (including panel members tested at high and low LoD). (F) %CV for TMB is shown. For (D) and (F), the red dotted line indicates acceptable %CV. Abbreviations: CNA: copy number amplification; CV: coefficient of variation; INDEL: insertion/deletion; LoD: limit of detection; MNV: multi-nucleotide variant; MSI-H: microsatellite instability high; MSS: microsatellite stable; SNV: single nucleotide variant; TMB: tumor mutational burden.
Laboratory developed test (LDT) validation
MI Cancer Seek is the FDA-approved version of its LDT predecessor, MI Tumor Seek Hybrid, which provides additional information relevant to indicated tumor types. While findings other than those listed in Table 1 for MI Cancer Seek are not prescriptive or conclusive for labeled use of any specific therapeutic product, these capabilities have been validated internally and meet CAP/CLIA requirements. Therefore, they may be considered at the physician’s discretion when evaluating possible treatments or clinical trials, or for research purposes. These LDT capabilities include the following: copy number variants (CNVs; including 1p19q co-deletion and 7+/10- co-occurrence), karyotype images, structural variants (SVs)/fusions, splice site variants (SSVs; including MET exon 14 skipping, ARv7, EGFRvIII), human leukocyte antigen (HLA) typing, loss of heterozygosity (LoH), homologous recombination deficiency (HRD) for predicting PARP inhibitor response in ovarian cancer, FOLFIRSTai signature for predicting first-line chemotherapy response in metastatic CRC, Genomic Probability Score (GPSai) for predicting tissue of origin in cancers of unknown primary (CUP), cancer-associated virus detection (HPV, EBV, MCPyV), and gene expression. Several of these capabilities have been or will be described in detail elsewhere, including HRD, FOLFIRSTai, and GPSai. The validation of HRD in ovarian cancer has been fully described [31]. Given the evolving landscape and potential clinical applications of this marker, HRD will also be reported for research-use-only on all tumor types, with further validations forthcoming. Development of the FOLFIRSTai 67-gene signature and the original GPSai model are described by Abraham et al. [32–34]. The GPSai model has since been updated and clinically validated, which has been reported separately. When compared to validated internal or external comparator assays (Supplementary Table 10), MI Tumor Seek Hybrid fulfilled acceptance criteria with regards to accuracy and precision for all LDT capabilities (Table 4).
Table 4: Concordance and precision of MI Tumor Seek Hybrid with comparator assays
LDT Capability | PPA (95% CI) accurate/total | NPA (95% CI) accurate/total | Precision (Average consensus call rate) |
---|---|---|---|
CNAa | 100% (93.7, 100) 57/57 | 99.9% (99.8, 100) 29,435/29,437 | 99.3% |
CNV lossb | 94.4% (86.6, 97.8) 68/72 | 98.9% (98.7, 99.0) 15,760/15,941 | 94.1% |
ERBB2 (HER2) CNA | 92.1% (89.2, 94.4) 409/444 | 99.6% (99.3, 99.8) 3,510/3,524 | N/A |
1p19q co-deletion | 100% (78.5, 100) 14/14 | 100% (85.7, 100) 23/23 | N/A |
7+/10- co-occurrence | 100% (78.5, 100) 14/14 | 96.2% (81.1, 99.3) 25/26 | N/A |
SV/fusion, SSVc | 98.2% (90.4, 99.7) 54/55 | N/Ad | SV/fusion: 93.0% |
ALK fusion | 94.3% (90.8, 96.8) 249/264 | 99.8% (99.7, 99.9) 8,932/8,949 | N/A |
HLA-A (other than A*2:01) | 99.3% (97.5, 99.8) 280/282 | N/A | 100% |
HLA-A*2:01 | 99.5% (97.3, 99.9) 201/202 | N/A | 100% |
HLA-B | 98.6% (97.0, 99.3) 477/484 | N/A | 100% |
HLA-C | 97.9% (96.2, 98.9) 474/484 | N/A | 100% |
LoH | 91.8% (82.2, 96.4) 56/61 | 92.3% (87.8, 95.2) 191/207 | 90.7% |
HRD (MI Exome) | 100% (70.1, 100) 9/9 | 100% (64.6, 100) 7/7 | 92.5% |
HRD (MyChoice) | 100% (51.0, 100) 4/4 | 100% (51.0, 100) 4/4 | |
FOLFIRSTai | 92.6% (76.6, 97.9) 25/27 | 92.9% (68.5, 98.7) 13/14 | N/A |
HPVe | 94.0% (83.8, 97.9) 47/50 | 99.4% (97.7, 99.8) 308/310 | 100% |
EBV | 100% (87, 100) 26/26 | 100% (87, 100) 26/26 | |
MCPYV | 100% (92.7, 100) 49/49 | 100% (84.5, 100) 21/21 |
Separate validations were performed in 3,968 samples for ERBB2 amplification in breast cancer as a complement to the MI Cancer Seek tumor profiling validation, and in 9,213 samples for ALK fusions in NSCLC. For this LDT validation, ERBB2 CNA results were compared to results of Ventana’s PATHWAY anti-HER2/neu (4B5) IHC test. These large-scale validations focusing on these clinically relevant alterations [35–38] in their respective tumor types yielded PPAs of 92.1% and 94.3% and NPAs of 99.6% and 99.8%, respectively (Table 4). An additional real-world analysis of MSI status in a cohort of 46,976 solid tumors and 3,488 endometrial tumors also demonstrated high concordance with the Ventana MMR RxDx Panel (Supplementary Table 11), further supporting our CDx claim.
Finally, MI Tumor Seek Hybrid provides gene expression data for panels of genes that are determined on a lineage basis for matching to clinical trials. Transcripts per million (TPM) and the accompanying percentile in the Caris cohort of the tumor type profiled are included in the report. Validation of MI Tumor Seek Hybrid gene expression data required a different approach due to the continuous nature of the data as well as technical differences from the comparator WTS assay, MI Transcriptome. We first determined area under the receiver operating characteristic curve (AUROC) for key genes that could be validated against immunohistochemistry (IHC). MI Tumor Seek Hybrid and MI Transcriptome demonstrated comparable AUROC values relative to pathologist-signed IHC results for androgen receptor (AR), HER2, estrogen receptor (ER), progesterone receptor (PR), and Claudin-18 (AUROC >0.89 for all) (Supplementary Figure 3). We then compared TPM between MI Transcriptome and MI Tumor Seek Hybrid numerically by applying a percentile transformation method, which is described in Supplementary Figure 4. Using this method, there was a strong correlation between MI Transcriptome and MI Tumor Seek Hybrid for a set of frequently reported genes (n = 214) (Supplementary Figure 5). Quantitative gene expression reporting by MI Tumor Seek Hybrid was also determined to be sufficiently precise, especially at TPM ≥25 (Supplementary Figure 6). Together, the gene expression validation studies support this LDT capability and culminate the comprehensive molecular reporting of MI Cancer Seek/MI Tumor Seek Hybrid.
DISCUSSION
We developed MI Cancer Seek (CDx) as a unique molecular assay designed to analyze both RNA and DNA simultaneously with a minimal DNA input of 50 ng and enhanced sequencing depth of genes recurrently altered in cancer. The technological workflow of MI Cancer Seek promotes efficient tissue utilization while providing comprehensive information on clinically actionable molecular vulnerabilities and additional clinical insights to be interpreted at the physician’s discretion. The results reported herein establish the clinical concordance of the MI Cancer Seek assay with appropriate comparator assays, including several FDA-approved tests. The CDx and tumor profiling claims for MI Cancer Seek are supplemented with relevant LDT capabilities including HRD, cancer-associated virus detection, and AI modeling. Together, the accuracy analysis substantially achieved or exceeded the acceptance criteria set forth, supporting MI Cancer Seek’s non-inferiority claims and CDx status.
Broad-based biomarker panels for precision medicine improve outcomes in a simplified, cost-effective manner [39–42] and are now recognized by major clinical oncology organizations such as ASCO as affording many advantages over more limited molecular testing [8]. Over the past several years, there has been an ever-expanding repertoire of newly FDA-approved targeted therapies or additional clinical indications for previously approved therapies with molecular biomarkers defining eligibility [8, 43–45], such as the tissue-agnostic approval of pembrolizumab for patients with MSI-high or TMB-high unresectable or metastatic solid tumors [46, 47]. Clinical trials testing various multi-modal therapy approaches including combinations of molecularly targeted therapies, immunotherapies, and chemotherapeutic agents further expand a physician’s biomarker-directed armamentarium [8, 48]. The importance of timely and accurate multi-gene testing to help guide these treatment decisions is underscored by data showing that that a large percentage of patients has at least one potentially actionable alteration [49, 50], while an absence of actionable alterations can help avoid unnecessary treatments that come with additional costs and side effects [51]. Multi-gene panel testing may also uncover reportable pathogenic variants in tumor tissue from patients not meeting criteria for hereditary genetic testing or may prompt eligibility for further germline testing [52–54].
Notwithstanding the advantages of multi-gene panels for oncology testing, multiple barriers exist that prevent their widespread clinical adoption, including lack of physician awareness and insufficient reimbursement for tests [43, 55]. Furthermore, inefficiencies in CDx testing such as poor test performance and poor sample quality or quantity may lead to significant loss in the oncology market and potentially severe repercussions for patients precluded from biomarker-matched therapy [43, 56]. Bearing these challenges in mind, integration of CDx tests into broad-based, multi-gene panels that optimize limited tissue resources is an essential step toward connecting as many patients as possible with personalized therapies [8].
Beyond the molecular test itself, a well-annotated and informative clinical report is essential for helping clinicians connect patients with molecularly-matched, FDA-approved, off-label, or trial therapies with varying levels of clinical consensus on the strength of the biomarker. A summary of reportable ranges and information included in the MI Cancer Seek clinical report is shown in Supplementary Table 12. Caris Life Sciences substantially adheres to and expands upon the joint consensus recommendations from the Association for Molecular Pathology, American College of Medical Genetics and Genomics (ACMG), ASCO, and CAP for essential elements of genomic test reports [57], including a tiered system for reporting the clinical significance of detected variants. Annotations are provided for each biomarker, with associated interpretive comments including functional, prognostic, predictive, and mechanistic data available from the literature. To enhance clarity and physician comprehension, cancer-type relevant biomarkers and associated therapies are highlighted, with all pathogenic and likely pathogenic variants subsequently reported. Whenever a relevant test fails due to issues with the sample (e.g., inadequate sample quantity or quality) or sequencing (e.g., insufficient depth), a notation is included in the report. While specific clinical trial recommendations are not made, a list of available clinical trials relevant to the identified biomarker is provided on the report. Clinicians also have access to the Clinical Trials Connector within their MI Portal interface, allowing them to perform real-time clinical trial searches based on location, biomarker, drugs, and trial sponsor. For appropriate patient groups, updates are provided in the form of a letter to the treating physician based on recent FDA drug approvals, further enhancing the potential impact of testing.
Collectively, the data in this validation indicate that the MI Cancer Seek assay is accurate, precise, and displays high analytical sensitivity and specificity. One limitation to be considered is the low PPA for ERBB2 CNA detection. While we demonstrated higher PPA for the comparison of ERBB2 CNA and HER2 IHC in our large LDT cohort (92.1%), the PPA for ERBB2 CNA versus FISH was suboptimal (84.7%). This difference in PPA may be the result of some discordance between FISH and IHC, which is known to occur particularly in weakly positive cases [58]. Thus, we suspect that MI Cancer Seek may identify cases with higher HER2 levels with greater accuracy than intermediate cases. Additionally, results from LDT capabilities of MI Tumor Seek Hybrid and any associated therapy recommendations are not to be taken as prescriptive or conclusive. Nonetheless, this assay demonstrates equivalency with orthogonal clinically validated tests and commercial, FDA-approved molecular tests, with the added advantage of simultaneous RNA and DNA analysis from the same minimal tissue input. In addition to providing an efficient molecular test option for patients/providers, the comprehensive molecular data produced by MI Cancer Seek/MI Tumor Seek Hybrid may also contribute to the compendium of tumor molecular information for research purposes. All components of the assay were validated by Caris under CAP/CLIA regulations for high-complexity laboratory testing, and it therefore represents an FDA-approved [20], efficient biomarker panel test that will contribute to the continued improvement and accessibility of precision medicine in oncology practice.
Materials and Methods
General methodology for validation
All samples, kits, reagents, and supplies were pre-specified and acquired in advance of the validation plan execution. All operators were pre-identified and trained on the workflow. All operators, samples, kit lot numbers, reagent lot numbers, expiration dates, instrument serial numbers, run dates, and validation results were documented. For any failures, inconsistencies, and/or questions, the technical supervisor (a board-certified molecular geneticist), and/or a board-certified pathologist was consulted. The studies were executed over multiple runs, by multiple operators and with different lots of reagents to ensure that testing performance was reproducible across multiple variables. The majority of cases for accuracy studies used an optimal input of DNA (220 ng), unless otherwise indicated. Non-optimal inputs (25, 37.5, 50, 275, and 330 ng) were tested for a DNA input and guard banding study. A summary of the study designs and parameters for all clinical and analytical studies performed is shown in Supplementary Table 13. Samples for the accuracy analysis underwent orthogonal testing using other Caris in-house assays or were tested using commercial assays by qualified laboratories (Supplementary Tables 2, 10).
Samples
A total of 2,063 unique samples, representative of 48 tumor lineages, were enrolled in the clinical validation studies. For clinical validation studies, biomarker positive samples were enrolled consecutively starting from the most recent case moving backward, and an equal number of date-matched biomarker negative samples were enrolled. Samples for the KRAS/NRAS study were enrolled consecutively regardless of biomarker status, since this study did not require biomarker enrichment. Samples were excluded from testing if they did not meet minimum requirements for MI Cancer Seek or the comparator assay. Samples with invalid results on MI Cancer Seek or the comparator assays were excluded from the primary analysis but were analyzed in best- and worst-case sensitivity analyses. For clinical validation studies, blinding was performed so that no internal or external operator was aware of the biomarker status of the tested sample. The demographics of the cohorts (age and sex) were comparable to the cohorts from the pivotal clinical trials investigating each biomarker. Samples for analytical validation studies shown here (e.g., DNA input, LoD, precision, interfering substances, LoB, cross-contamination, carry-over) were selected based on sample biomarker status and where applicable were diluted with normal tissue samples or other cancer samples as indicated to achieve designated DNA input levels or tumor purity, VF, etc. according to prespecified study designs. Additional samples were used for the tumor profiling validation, analytical validation, and LDT validation studies (Supplementary Table 13). Positive and negative controls specific to each capability of the assay were included in this study and are included for each MI Cancer Seek run.
Total nucleic acid (TNA) extraction
FFPE slides underwent review by a board-certified pathologist to measure tumor content. For accuracy analyses, a minimum of 20% tumor content across 25 mm2 area was required, with manual microdissection performed to meet requirements if necessary. Microdissection was also performed as needed to exclude interfering substances as much as possible (melanin, adipose, necrosis). TNA was auto-extracted using a MagMax FFPE DNA/RNA Ultra extraction kit (Thermo Fisher Scientific, Waltham, MA, USA) on a Biomek i7 Automated Workstation (Beckman Coulter, Brea, CA, USA) and KingFisher Flex (Thermo Fisher Scientific). DNA quantification was performed using a Quant-iT 1X dsDNA High Sensitivity Assay Kit (Thermo Fisher Scientific) and read on a GloMax® Microplate Reader (Promega, Madison, WI, USA). Samples were prepared for quantification, normalized, and plated for downstream processes using a Biomek i7 Automated Workstation. RNA quantification was not required, since starting DNA and RNA molecules were converted into libraries for NGS in the same hybrid workflow. The RNA/DNA ratio in TNA from tissue was always >1; thus, the input into library preparation was controlled by limiting material (DNA).
MI Cancer Seek library preparation and next-generation sequencing
Library preparation was automated on the Bravo B Liquid Handler (Agilent, Santa Clara, CA, USA) using KAPA NGS Library preparation reagents (Roche Diagnostics, Indianapolis, IN, USA) and custom cDNA primers (IDT, Coralville, IA, USA; GeneLink, Elmsford, NY, USA). NGS library preparation is a fully customized workflow, referred to as a ‘hybrid’ workflow, where starting TNA molecules proceed through the process together. DNA and RNA underwent fragmentation, followed by cDNA synthesis from RNA, end repair and A-tailing of cDNA and original DNA, ligation of universal adapters, pre-capture PCR, and addition of P5/P7 indexes. Genomic targets were enriched through hybridization with a custom bait panel and captured using 120 nucleotides long, double-stranded, biotinylated baits. The bait panel was customized to add additional coverage for clinically relevant and reportable genes (Supplementary Table 6). Capture and purification of enriched DNA targets was performed following hybridization of dsDNA probes (baits) to adapter-ligated DNA. Post-capture PCR was performed on enriched DNA targets, and the resulting library was quantified using a Quant-iT 1X dsDNA High Sensitivity Assay Kit. Finally, normalization and pooling were performed, with quantification of the final library pool by a Qubit 1X dsDNA High Sensitivity Assay Kit (Thermo Fisher Scientific). Sequencing was performed on a NovaSeq 6000 instrument (Illumina, San Diego, CA, USA), using recommended reagents. A Veriti Thermocycler (Thermo Fisher Scientific) was used for all PCR steps. Quality control elements were checked at applicable steps. RNA was chemically labeled during library preparation, which enables separating RNA-originated data during bioinformatics data analysis. The MI Cancer Seek workflow is shown in Figure 1.
MI Cancer Seek bioinformatic pipeline
Sequencing data were extracted into split FASTQ files (RNA and DNA) for further processing through Caris’ proprietary bioinformatics pipeline. For RNA, Spliced Transcripts Alignment to a Reference (STAR) software was used for alignment, and STAR-Fusion was employed for fusion detection [59]. Transcripts per million (TPM) molecules were generated using the Salmon expression pipeline [60]. DNA variants detected were mapped to reference genome hg38, and well-established bioinformatics tools such as BWA, Samtools, Pindel, and snpEff were incorporated to perform variant calling functions; germline variants were filtered with various germline databases, including dbSNP. CNVs were detected using CNVkit [61]. Genetic variants identified were interpreted by board-certified molecular geneticists and annotated as ‘pathogenic,’ ‘likely pathogenic,’ ‘variant of unknown significance,’ ‘likely benign,’ or ‘benign,’ according to ACMG standards.
Tumor signatures
The threshold to determine MSI-high status was 39 or more loci with frameshift mutations out of 5,721 loci examined. TMB was measured by counting all non-synonymous missense, nonsense, in-frame insertion/deletion, and frameshift mutations found per tumor that had not been previously described as germline alterations in dbSNP151, Genome Aggregation Database (gnomAD, AC>0) databases, or benign variants identified by Caris geneticists. A cutoff point of ≥10 mutations per MB was used based on the KEYNOTE-158 pembrolizumab trial [62].
LoH was determined by calculating the LoH of single nucleotide polymorphisms (SNPs) within 552 chromosome segments of 2–6 Mb in length. 250K SNPs were analyzed, with a minimum of 17 SNPs per Mb of genome sequence. The final call of genomic LoH was based on the percentage of all 552 segments with observed LoH (High ≥16%; Equivocal ≥11% and ≤15%; Low <11%; Indeterminate ≤3,000 SNPs read or sample depth <200×). A normal epithelial ovarian genome (NA12878) that has no non-polymorphic variants, gene fusions or other cancer hallmarks, was used as a negative control. LoH was also used to determine HRD. The presence of LoH and large-scale state transitions (LST), which are the loss of chromosomal segments larger than 10 Mb, contributed to the genomic scar score (GSS). HRD was called when the sample was positive for BRCA1 or BRCA2 mutation or was genomic scar score (GSS)-high (GSS ≥46), and negative when it did not meet those criteria. HRD was restricted to ovarian cancer cases.
Statistical analysis
Sample sizes for CDx studies were targeted to achieve at least 80% power to rule out non-inferiority, accounting for projected invalid rates based on historical data. The agreement between two replicates of the comparator companion diagnostic (CCD1 and CCD2) was compared to the agreements of the follow-on companion diagnostic (FCD) to CCD1 and FCD to CCD2, according to Li et al. where a reference standard is not available and the concordance study sample is not a random sample from the MI Cancer Seek intended use population (with the exception of KRAS/NRAS, since no enrichment was required) [28]. Concordance results were used to calculate agreement values (NPA and PPA) and differences (𝜁) in those values. Differences in agreements were calculated according to the following formulas: 𝜁PPA1 = PPA C1C2 − PPAC1F; 𝜁PPA2 = PPA C2C1 − PPAC2F; 𝜁NPA1 = NPA C1C2 − NPAC1F; and 𝜁NPA2 = NPA C2C1 − NPAC2F. NPA/PPAC1C2 is the proportion of CCD1 negative/positive results in which CCD2 is negative/positive. NPA/PPAC1F is the proportion of CCD1 negative/positive results in which FCD is negative/positive. NPA/PPAC2C1 is the proportion of CCD2 negative/positive results in which CCD1 is negative/positive. NPA/PPAC2F is the proportion of CCD2 negative/positive results in which FCD is negative/positive. Prevalence-adjusted agreement values (i.e. adjusted for enrichment of the sample population) were calculated using Bayes’ Theorem and the Law of Total Probability.
FCD was considered non-inferior to CCD by a margin of ε, where ε is the maximum upper limit of the 95% CIs for each 𝜁, and δ is the target non-inferiority margin. ε1 is the true non-inferiority margin for FCD as a predictive test, ε0 is the true non-inferiority margin for FCD as a selective test, δ1 is the target non-inferiority margin for FCD as a predictive test, and δ0 is the target non-inferiority margin for FCD as a selective test. If ε <δ, FCD achieved the targeted non-inferiority level. Two-sided 95% confidence intervals (CIs) for agreement differences (𝜁) were calculated using the percentile bootstrap method with 1000 bootstrap samples for all studies except KRAS/NRAS, which used the continuity-corrected Wilson interval for paired data, as described in method 10 of Newcombe et al. [63]. Sensitivity analyses were performed to evaluate the impact of prevalence using an upper and lower bound based on values reported in the literature. Standard approaches in JMP Version 17 or higher, SAS Version 9.4 or higher, or R version 4.3.0 or higher were used for data analysis.
Abbreviations
ACMG: American College of Medical Genetics and Genomics; AR: androgen receptor; ASCO: American Society of Clinical Oncology; AUROC: area under the receiver operating characteristic curve; CAP: College of American Pathologists; CCD: comparator companion diagnostic; CDx: companion diagnostic; CI: confidence interval; CLIA: Clinical Laboratory Improvement Amendments; CNA: copy number amplification; CNV: copy number variation; CRC: colorectal cancer; CUP: cancer of unknown primary; ER: estrogen receptor; ESMO: European Society for Medical Oncology; FCD: follow-on companion diagnostic; FDA: Food and Drug Administration; FFPE: formalin-fixed paraffin-embedded; GPS: genomic probability score; GSS: genomic scar score; IHC: immunohistochemistry; INDEL: insertion/deletion; LDT: laboratory developed test; LoB: limit of blank; LoD: limit of detection; LST: large-scale state transitions; MI: Molecular Intelligence; MSI: microsatellite instability; MSS: microsatellite stable; NCCN: National Comprehensive Cancer Network; NGS: next-generation sequencing; NPA: negative percent agreement; NSCLC: non-small cell lung cancer; PPA: positive percent agreement; PR: progesterone receptor; SNV: single-nucleotide polymorphism; TMB: tumor mutational burden; TNA: total nucleic acid; TPM: transcripts per million; VF: variant frequency; WES: whole exome sequencing; WT: wild-type; WTS: whole transcriptome sequencing.
Data availability
The Caris Life Sciences database is a proprietary database; therefore, the raw molecular data produced throughout this study cannot be publicly shared. Biomarker results and agreements are shown in full in the main figures/tables and supplementary files. For further information, please contact Dr. David Spetzler at
ACKNOWLEDGMENTS
We thank all Caris Life Sciences employees who contributed to the development and launch of MI Cancer Seek.
CONFLICTS OF INTEREST
All authors have a financial relationship as employees of Caris Life Sciences.
ETHICAL STATEMENT AND CONSENT
This study was conducted in accordance with guidelines of the Declaration of Helsinki, Belmont report, and U.S. Common Rule. All samples (aside from a small number of samples commercially purchased for limit of blank study) were selected from the Caris Life Sciences remnant biorepository, which has an exempt determination under 45 CFR § 46.104(d)(4), issued by WCG Institutional Review Board (IRB). Protocols for non-inferiority and tumor profiling validation testing were individually reviewed per 21 CFR 56, and were approved by WCG IRB with waivers of patient informed consent. All samples were deidentified and remained deidentified for all testing. No protected health information was used or released during any testing.
FUNDING
No external funding was received for this study.
References
1. Fernandez-Rozadilla C, Simões AR, Lleonart ME, Carnero A, Carracedo Á. Tumor Profiling at the Service of Cancer Therapy. Front Oncol. 2021; 10:595613. https://doi.org/10.3389/fonc.2020.595613. [PubMed].
2. Isaak AJ, Clements GR, Buenaventura RGM, Merlino G, Yu Y. Development of Personalized Strategies for Precisely Battling Malignant Melanoma. Int J Mol Sci. 2024; 25:5023. https://doi.org/10.3390/ijms25095023. [PubMed].
3. Piombino C, Pipitone S, Tonni E, Mastrodomenico L, Oltrecolli M, Tchawa C, Matranga R, Roccabruna S, D’Agostino E, Pirola M, Bacchelli F, Baldessari C, Baschieri MC, et al. Homologous Recombination Repair Deficiency in Metastatic Prostate Cancer: New Therapeutic Opportunities. Int J Mol Sci. 2024; 25:4624. https://doi.org/10.3390/ijms25094624. [PubMed].
4. Garg P, Krishna M, Subbalakshmi AR, Ramisetty S, Mohanty A, Kulkarni P, Horne D, Salgia R, Singhal SS. Emerging biomarkers and molecular targets for precision medicine in cervical cancer. Biochim Biophys Acta Rev Cancer. 2024; 1879:189106. https://doi.org/10.1016/j.bbcan.2024.189106. [PubMed].
5. Milbury CA, Creeden J, Yip WK, Smith DL, Pattani V, Maxwell K, Sawchyn B, Gjoerup O, Meng W, Skoletsky J, Concepcion AD, Tang Y, Bai X, et al. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors. PLoS One. 2022; 17:e0264138. https://doi.org/10.1371/journal.pone.0264138. [PubMed].
6. Beaubier N, Tell R, Lau D, Parsons JR, Bush S, Perera J, Sorrells S, Baker T, Chang A, Michuda J, Iguartua C, MacNeil S, Shah K, et al. Clinical validation of the tempus xT next-generation targeted oncology sequencing assay. Oncotarget. 2019; 10:2384–96. https://doi.org/10.18632/oncotarget.26797. [PubMed].
7. Muquith M, Espinoza M, Elliott A, Xiu J, Seeber A, El-Deiry W, Antonarakis ES, Graff SL, Hall MJ, Borghaei H, Hoon DSB, Liu SV, Ma PC, et al. Tissue-specific thresholds of mutation burden associated with anti-PD-1/L1 therapy benefit and prognosis in microsatellite-stable cancers. Nat Cancer. 2024; 5:1121–29. https://doi.org/10.1038/s43018-024-00752-x. [PubMed].
8. Chakravarty D, Johnson A, Sklar J, Lindeman NI, Moore K, Ganesan S, Lovly CM, Perlmutter J, Gray SW, Hwang J, Lieu C, André F, Azad N, et al. Somatic Genomic Testing in Patients With Metastatic or Advanced Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol. 2022; 40:1231–58. https://doi.org/10.1200/JCO.21.02767. [PubMed].
9. Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, Normanno N, Scarpa A, Robson M, Meric-Bernstam F, Wagle N, Stenzinger A, Bonastre J, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020; 31:1491–505. https://doi.org/10.1016/j.annonc.2020.07.014. [PubMed].
10. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: colorectal, Version 3. 2024. https://www.nccn.org/professionals/physician_gls/pdf/genetics_ceg.pdf.
11. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, and Prostate, Version 2. 2025. https://www.nccn.org/professionals/physician_gls/pdf/genetics_bopp.pdf.
12. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Pancreatic Adenocarcinoma, Version 1. 2025. https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf.
13. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Breast Cancer, Version 6. 2024. https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf.
14. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Colon Cancer Version 5. 2024. https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf.
15. Dotolo S, Esposito Abate R, Roma C, Guido D, Preziosi A, Tropea B, Palluzzi F, Giacò L, Normanno N. Bioinformatics: From NGS Data to Biological Complexity in Variant Detection and Oncological Clinical Practice. Biomedicines. 2022; 10:2074. https://doi.org/10.3390/biomedicines10092074. [PubMed].
16. Singh RR. Next-Generation Sequencing in High-Sensitive Detection of Mutations in Tumors: Challenges, Advances, and Applications. J Mol Diagn. 2020; 22:994–7. https://doi.org/10.1016/j.jmoldx.2020.04.213. [PubMed].
17. Deans ZC, Costa JL, Cree I, Dequeker E, Edsjö A, Henderson S, Hummel M, Ligtenberg MJ, Loddo M, Machado JC, Marchetti A, Marquis K, Mason J, et al, and IQN Path ASBL. Integration of next-generation sequencing in clinical diagnostic molecular pathology laboratories for analysis of solid tumours; an expert opinion on behalf of IQN Path ASBL. Virchows Arch. 2017; 470:5–20. https://doi.org/10.1007/s00428-016-2025-7. [PubMed].
18. Astolfi A, Urbini M, Indio V, Nannini M, Genovese CG, Santini D, Saponara M, Mandrioli A, Ercolani G, Brandi G, Biasco G, Pantaleo MA. Whole exome sequencing (WES) on formalin-fixed, paraffin-embedded (FFPE) tumor tissue in gastrointestinal stromal tumors (GIST). BMC Genomics. 2015; 16:892. https://doi.org/10.1186/s12864-015-1982-6. [PubMed].
19. Rusch M, Nakitandwe J, Shurtleff S, Newman S, Zhang Z, Edmonson MN, Parker M, Jiao Y, Ma X, Liu Y, Gu J, Walsh MF, Becksfort J, et al. Clinical cancer genomic profiling by three-platform sequencing of whole genome, whole exome and transcriptome. Nat Commun. 2018; 9:3962. https://doi.org/10.1038/s41467-018-06485-7. [PubMed].
20. United States Food and Drug Administration. Premarket Approval (PMA) MI Cancer Seek. 2024.
21. Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, Wasan H, Ciardiello F, Loupakis F, Hong YS, Steeghs N, Guren TK, Arkenau HT, et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N Engl J Med. 2019; 381:1632–43. https://doi.org/10.1056/NEJMoa1908075. [PubMed].
22. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, et al, and Spanish Lung Cancer Group in collaboration with Groupe Français de Pneumo-Cancérologie and Associazione Italiana Oncologia Toracica. 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; 13:239–46. https://doi.org/10.1016/S1470-2045(11)70393-X. [PubMed].
23. André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, Iwata H, Conte P, Mayer IA, Kaufman B, Yamashita T, Lu YS, Inoue K, et al, and SOLAR-1 Study Group. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N Engl J Med. 2019; 380:1929–40. https://doi.org/10.1056/NEJMoa1813904. [PubMed].
24. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD. Wild-Type KRAS Is Required for Panitumumab Efficacy in Patients With Metastatic Colorectal Cancer. J Clin Oncol. 2023; 41:3278–86. https://doi.org/10.1200/JCO.22.02758. [PubMed].
25. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N, Kudchadkar R, Burris HA 3rd, Falchook G, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012; 367:1694–703. https://doi.org/10.1056/NEJMoa1210093. [PubMed].
26. Maio M, Ascierto PA, Manzyuk L, Motola-Kuba D, Penel N, Cassier PA, Bariani GM, De Jesus Acosta A, Doi T, Longo F, Miller WH, Oh DY, Gottfried M, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol. 2022; 33:929–38. https://doi.org/10.1016/j.annonc.2022.05.519. [PubMed].
27. Makker V, Colombo N, Casado Herráez A, Monk BJ, Mackay H, Santin AD, Miller DS, Moore RG, Baron-Hay S, Ray-Coquard I, Ushijima K, Yonemori K, Kim YM, et al. Lenvatinib Plus Pembrolizumab in Previously Treated Advanced Endometrial Cancer: Updated Efficacy and Safety From the Randomized Phase III Study 309/KEYNOTE-775. J Clin Oncol. 2023; 41:2904–10. https://doi.org/10.1200/JCO.22.02152. [PubMed].
28. Li M. Statistical Methods for Clinical Validation of Follow-On Companion Diagnostic Devices via an External Concordance Study. Stat Biopharm Res. 2016; 8:355–63.
29. Personal Genome Diagnostics. PGDx elioTM tissue complete product brochure. 2024. https://www.personalgenome.com/uploads/downloads/19987_BRO_PGDxeliotissuecomplete.pdf.
30. Keefer LA, White JR, Wood DE, Gerding KMR, Valkenburg KC, Riley D, Gault C, Papp E, Vollmer CM, Greer A, Hernandez J, McGregor PM 3rd, Zingone A, et al. Automated next-generation profiling of genomic alterations in human cancers. Nat Commun. 2022; 13:2830. https://doi.org/10.1038/s41467-022-30380-x. [PubMed].
31. Evans E, Dholakia J, Abraham J, Hinton A, Zhang J, Xiu J, Maney T, Oberley M, Thacker P, Herzog TJ, Spetzler D, Arend RC. Whole-exome sequencing provides assessment of homologous recombination deficiency for identification of PARPi-responsive ovarian tumors. Gynecol Oncol. 2025; 197:139–45. https://doi.org/10.1016/j.ygyno.2025.04.586. [PubMed].
32. Abraham JP, Magee D, Cremolini C, Antoniotti C, Halbert DD, Xiu J, Stafford P, Berry DA, Oberley MJ, Shields AF, Marshall JL, Salem ME, Falcone A, et al. Clinical Validation of a Machine-learning-derived Signature Predictive of Outcomes from First-line Oxaliplatin-based Chemotherapy in Advanced Colorectal Cancer. Clin Cancer Res. 2021; 27:1174–83. https://doi.org/10.1158/1078-0432.CCR-20-3286. [PubMed].
33. Abraham J, Heimberger AB, Marshall J, Heath E, Drabick J, Helmstetter A, Xiu J, Magee D, Stafford P, Nabhan C, Antani S, Johnston C, Oberley M, et al. Machine learning analysis using 77,044 genomic and transcriptomic profiles to accurately predict tumor type. Transl Oncol. 2021; 14:101016. https://doi.org/10.1016/j.tranon.2021.101016. [PubMed].
34. Ghani H, Helmstetter A, Ribeiro JR, Maney T, Rock S, Feldman RA, Swensen J, Abdulla F, Spetzler DB, Florento E, Vanderwalde AM, Pittman P, Radovich M, et al. GPSai: A Clinically Validated AI Tool for Tissue of Origin Prediction During Routine Tumor Profiling. Cancer Res Commun. 2025. [Epub ahead of print]. https://doi.org/10.1158/2767-9764.CRC-25-0171. [PubMed].
35. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001; 344:783–92. https://doi.org/10.1056/NEJM200103153441101. [PubMed].
36. Shaw AT, Gandhi L, Gadgeel S, Riely GJ, Cetnar J, West H, Camidge DR, Socinski MA, Chiappori A, Mekhail T, Chao BH, Borghaei H, Gold KA, et al, and study investigators. Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: a single-group, multicentre, phase 2 trial. Lancet Oncol. 2016; 17:234–42. https://doi.org/10.1016/S1470-2045(15)00488-X. [PubMed].
37. Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn MJ, De Pas T, Besse B, Solomon BJ, Blackhall F, Wu YL, Thomas M, O’Byrne KJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013; 368:2385–94. https://doi.org/10.1056/NEJMoa1214886. [PubMed].
38. Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, Solomon BJ, Wolf J, et al. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014; 370:1189–97. https://doi.org/10.1056/NEJMoa1311107. [PubMed].
39. Schwaederle M, Parker BA, Schwab RB, Daniels GA, Piccioni DE, Kesari S, Helsten TL, Bazhenova LA, Romero J, Fanta PT, Lippman SM, Kurzrock R. Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience. Mol Cancer Ther. 2016; 15:743–52. https://doi.org/10.1158/1535-7163.MCT-15-0795. [PubMed].
40. Brito RA, Cullum B, Hastings K, Avalos-Reyes E, Karos R, Johnson KA. Total cost of lung cancer care associated with broad panel versus narrow panel sequencing. J Clin Oncol. 2020; 38:7077. https://doi.org/10.1200/JCO.2020.38.15_suppl.7077.
41. Hofmarcher T, Malmberg C, Lindgren P. A global analysis of the value of precision medicine in oncology - The case of non-small cell lung cancer. Front Med (Lausanne). 2023; 10:1119506. https://doi.org/10.3389/fmed.2023.1119506. [PubMed].
42. Kato S, Kim KH, Lim HJ, Boichard A, Nikanjam M, Weihe E, Kuo DJ, Eskander RN, Goodman A, Galanina N, Fanta PT, Schwab RB, Shatsky R, et al. Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy. Nat Commun. 2020; 11:4965. https://doi.org/10.1038/s41467-020-18613-3. [PubMed].
43. Keeling P, Clark J, Finucane S. Challenges in the clinical implementation of precision medicine companion diagnostics. Expert Rev Mol Diagn. 2020; 20:593–99. https://doi.org/10.1080/14737159.2020.1757436. [PubMed].
44. United States Food and Drug Administration. GUIDANCE DOCUMENT: Tissue Agnostic Drug Development in Oncology, Draft Guidance for Industry. 2022. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/tissue-agnostic-drug-development-oncology.
45. Dou YN, Grimstein C, Mascaro J, Wang J. Biomarkers for Precision Patient Selection in Cancer Therapy Approvals in the US, from 2011 to 2023. Clin Pharmacol Ther. 2024; 116:304–14. https://doi.org/10.1002/cpt.3306. [PubMed].
46. Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin Cancer Res. 2019; 25:3753–58. https://doi.org/10.1158/1078-0432.CCR-18-4070. [PubMed].
47. United States Food and Drug Administration. FDA approves pembrolizumab for adults and children with TMB-H solid tumors. 2020. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors.
48. Coyne GO, Takebe N, Chen AP. Defining precision: The precision medicine initiative trials NCI-MPACT and NCI-MATCH. Curr Probl Cancer. 2017; 41:182–93. https://doi.org/10.1016/j.currproblcancer.2017.02.001. [PubMed].
49. Schwaederle M, Daniels GA, Piccioni DE, Fanta PT, Schwab RB, Shimabukuro KA, Parker BA, Kurzrock R. On the Road to Precision Cancer Medicine: Analysis of Genomic Biomarker Actionability in 439 Patients. Mol Cancer Ther. 2015; 14:1488–94. https://doi.org/10.1158/1535-7163.MCT-14-1061. [PubMed].
50. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, Chakravarty D, Daian F, Gao Q, et al, and Cancer Genome Atlas Research Network. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018; 173:321–37.e10. https://doi.org/10.1016/j.cell.2018.03.035. [PubMed].
51. Mathur S, Sutton J. Personalized medicine could transform healthcare. Biomed Rep. 2017; 7:3–5. https://doi.org/10.3892/br.2017.922. [PubMed].
52. Lincoln SE, Nussbaum RL, Kurian AW, Nielsen SM, Das K, Michalski S, Yang S, Ngo N, Blanco A, Esplin ED. Yield and Utility of Germline Testing Following Tumor Sequencing in Patients With Cancer. JAMA Netw Open. 2020; 3:e2019452. https://doi.org/10.1001/jamanetworkopen.2020.19452. [PubMed].
53. Meric-Bernstam F, Brusco L, Daniels M, Wathoo C, Bailey AM, Strong L, Shaw K, Lu K, Qi Y, Zhao H, Lara-Guerra H, Litton J, Arun B, et al. Incidental germline variants in 1000 advanced cancers on a prospective somatic genomic profiling protocol. Ann Oncol. 2016; 27:795–800. https://doi.org/10.1093/annonc/mdw018. [PubMed].
54. Mandelker D, Zhang L, Kemel Y, Stadler ZK, Joseph V, Zehir A, Pradhan N, Arnold A, Walsh MF, Li Y, Balakrishnan AR, Syed A, Prasad M, et al. Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing. JAMA. 2017; 318:825–35. https://doi.org/10.1001/jama.2017.11137. [PubMed].
55. Schroll MM, Agarwal A, Foroughi O, Kong E, Perez O, Pritchard D, Breur T, Garner Amanti K, Gustavsen G. Stakeholders Perceptions of Barriers to Precision Medicine Adoption in the United States. J Pers Med. 2022; 12:1025. https://doi.org/10.3390/jpm12071025. [PubMed].
56. Clark J, Smart D, Keeling P. Patients lost to precision treatment due to suboptimal testing: US and EU real world evidence. European Alliance for Personalised Medicine Congress (EAPM). Belfast City, North Ireland. 2017.
57. Li MM, Datto M, Duncavage EJ, Kulkarni S, Lindeman NI, Roy S, Tsimberidou AM, Vnencak-Jones CL, Wolff DJ, Younes A, Nikiforova MN. Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer: A Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017; 19:4–23. https://doi.org/10.1016/j.jmoldx.2016.10.002. [PubMed].
58. Zhao B, Wang Y, Xu H. Concordance of immunohistochemistry and fluorescence in situ hybridization for assessment of HER2 status in breast cancer patients in Xinjiang autonomous region, China. Int J Clin Exp Pathol. 2017; 10:10459–66. [PubMed].
59. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29:15–21. https://doi.org/10.1093/bioinformatics/bts635. [PubMed].
60. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017; 14:417–19. https://doi.org/10.1038/nmeth.4197. [PubMed].
61. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS Comput Biol. 2016; 12:e1004873. https://doi.org/10.1371/journal.pcbi.1004873. [PubMed].
62. Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, Chung HC, Kindler HL, Lopez-Martin JA, Miller WH Jr, Italiano A, Kao S, Piha-Paul SA, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020; 21:1353–65. https://doi.org/10.1016/S1470-2045(20)30445-9. [PubMed].
63. Newcombe RG. Improved confidence intervals for the difference between binomial proportions based on paired data. Stat Med. 1998; 17:2635–50. [PubMed].