International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Comprehensive genomic profiling (CGP) is a molecular diagnostic tool, based on next-generation sequencing (NGS) technology, used in the field of cancer research and treatment. CGP identifies various genetic alterations and mutations within the tumor (1). Results from CGP guide oncologists to make decisions about treatment options and select targeted therapies tailored to the specific genetic alterations present within the tumor. CGP examines a broad panel of genes, detecting a wide range of genomic alterations. This includes point mutations (such as PIK3CA p.E453Q and c.1357G>C), insertions/deletions (indels; such as BRCA2 p.E1571Gfs*3 and c.4712_4713delAG), copy number variations (CNVs; such as ERBB2 amplification, copy number:4) and gene fusions (such as TBL1XR1/PIK3CA fusion). It can detect multiple genetic alterations in a single test, reducing the need for multiple individual tests. CGP is an application of NGS that specifically focuses on a detailed analysis of the genetic profile of patients, and can provide a broad overview of the genomic alterations in a tumor. Consequently, precision, efficiency and therapeutic guidance are potential advantages of CGP, which has been advocated for advanced-stage cancers (2-5). A large-sized panel can provide more opportunities for matching patients to targeted therapies or for increased mutation detection in clinical trials, while the use of CGP is limited by its high cost, data complexity and increased turnaround time.
Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer characterized by the absence of three deterministic receptors commonly found in other breast cancers: Estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2). TNBC cells lack the expression of these three receptors and do not rely on them for growth. TNBC accounts for 10-20% of all breast cancer cases world (6), and is more commonly diagnosed in younger (#x003C;40 years old) women, African-American women and those with a family history of breast cancer (7). TNBC is known for its aggressive behavior and tends to grow and spread quickly, while it is also associated with a higher risk of recurrence and metastasis (8), and ~40% of people with stage I to stage III TNBC will exhibit tumor recurrence after standard treatment (9). Treatment options for TNBC often involve chemotherapy, as endocrine therapies and anti-HER2 therapies that target ER, PR or HER2 are not effective due to the absence of these receptors (8). The prognosis for TNBC varies depending on factors such as the diagnosed stage and response to treatment (10). Due to its aggressiveness, there is an unmet need to identify actionable targets such as AKT1, BRCA1/2, PALB2, ERBB2, PIK3CA and PTEN, for patients with TNBC (11,12).
Research focusing on developing targeted therapies for TNBC continues to improve treatment outcomes and broaden therapeutic options (13,14). For example, biomarker-driven therapies are being applied with the advent of immune checkpoint inhibitors, poly-adenosine-diphosphate-ribose polymerase (PARP) inhibitors, selective PI3K and AKT inhibitors, as well as antibody-drug conjugates, which target immune-enriched, DNA repair-deficient, PI3K/AKT/mTOR-activated and surface antigen-overexpressed TNBC, respectively. However, most umbrella trials for advanced breast cancer are not TNBC-specific, while a recent review highlighted that large-sized panels could identify novel markers, emphasizing the potential of CGP to uncover actionable targets in TNBC (3).
Commercialized CGP assays may be beneficial; however, the size of the panel to be employed to deliver the most optimal coverage of actionable genes for TNBC remains to be explored. Some studies comparing distinct CGP platforms and resulting biomarkers have been conducted for gastric, head and neck squamous cell carcinoma, non-small cell lung cancer, ovarian and prostate cancer, but rarely for TNBC (15,16). In the present study, a subgroup of patients with TNBC from the Veterans General Hospital TAipei-Yung-Ling foundation sinO-canceR (VGH-TAYLOR) study was used (17). TNBC samples were initially assayed using a medium-sized CGP panel, and the remaining specimens of nucleic acid were re-sequenced with a large-sized GCP panel. The aim of the present study was to investigate whether a larger CGP panel offers clinically significant advantages in detecting actionable variants for TNBC management compared with a medium-sized panel.
The present study was conducted in two phases: The first phase involved prospective and retrospective tissue collection as part of the VGH-TAYLOR study (medium-sized panel). The second phase involved the reuse of biobanked samples from the VGH-TAYLOR study (large-sized panel). The study was approved by the Institutional Review Board of Taipei Veterans General Hospital (approval nos. 2021-01-007B and 2023-08-002B; Taipei, Taiwan). Participants provided written informed consent for both phases of the study.
The VGH-TAYLOR study was designed to examine the genetic profiling of different subtypes of breast cancer in Taiwan (17). The prospective study comprised diverse clinical scenarios of breast cancer: i) Group 1, planned to receive first-line surgery followed by adjuvant therapy (1A) or exhibited early relapse within 3 years of diagnosis (1B); ii) group 2, planned to receive first-line neoadjuvant therapy followed by surgery; and iii) group 3, exhibited de novo stage IV (3-I) or recurrence beyond 3 years of diagnosis (3-II).
In addition, a retrospective cohort with biobanked samples from recurrent/metastatic breast cancers or patients with non-pathological complete response following neoadjuvant therapy was also included (18-21).
Inclusion criteria were a diagnosis of TNBC from the VGH-TAYLOR study, availability of sufficient biobanked nucleic acid and willingness to sign informed consent. Patients without residual samples and those unwilling to participate in the retesting program were excluded. A total of 120 female patients were screened, with 108 cases included for comprehensive genomic profiling. The age range was 27 to 84 years, with a mean ± SD age of 56.1±12.8 years. All enrolled subjects were treated according to contemporary guidelines and underwent regular follow-up (22,23). Regarding immunohistochemistry (IHC) testing, ER and PR were scored by percentage of nuclear labeling (0-100%). HER2 expression was scored using a 0 to 3+ membrane staining intensity score (24). Hormone receptor positivity was defined by either ER or PR with ≥1% of tumor cells exhibiting nuclear staining while HR-negative breast cancer was defined by the absence of ER and PR on the cancer cells. HER2 overexpression was indicated by either a 3+ (positive) or 2+ (equivocal) IHC score with fluorescence in situ hybridization (FISH) amplification (25). For patients with equivocal IHC scoring of HER2, a negative FISH result was a prerequisite. All patients of the VGH-TAYLOR study were enrolled between November 2018 and December 2021 from the Comprehensive Breast Health Center, Taipei Veterans General Hospital, a tertiary referral medical center at Taipei, Taiwan.
Formalin-fixed paraffin-embedded (FFPE) samples were collected after obtaining informed consent. For consistent results, a fixation time of 6-48 h in 10% neutral buffer formalin at room temperature for breast cancer biomarker testing was performed. At least seven unstained tumor sections with 10 µm in thickness were retrieved, with one used for hematoxylin and eosin (H&E) staining and six used for nucleic acid extraction. Hematoxylin (3.5 min at room temperature) was used for nuclear staining, and eosin (60 sec at room temperature) was used for cytoplasmic staining, allowing for differentiation of cellular components. H&E-stained slides were reviewed to ascertain the presence of adequate breast cancer cells (>70% of cancer composition).
Paraffin was removed by xylene and ethanol serial washes. Nucleic acid was extracted from 5-µm sections with the QIAmp DNA FFPE Tissue Kit (cat. no. 56404; Qiagen, Inc.) or AllPrep DNA/RNA FFPE Kit (cat. no. 80234; Qiagen, Inc.), while quality control and concentration were checked and determined using the Qubit fluorimeter (Invitrogen; Thermo Fisher Scientific, Inc.), Qubit dsDNA HS and Qubit dsDNA BR Assay Kits (cat. nos. Q32851 and Q32850; Thermo Fisher Scientific, Inc.). In the current study, treatment-naïve cancerous tissue was used for NGS. Targeted sequencing for the TruSight Oncology 500 (TSO500) panel was conducted on residual specimens from the VGH-TAYLOR study.
The storage conditions for the remaining nucleic acids after CGP aimed at maintaining their integrity for potential future use. Therefore, nuclear acid was stored at -80˚C in Tris-EDTA buffer or nuclease-free water. Repeated freeze-thaw cycles were avoided. A total of 120 pairs of DNA/RNA were tested for integrity and 108 passed quality control (QC) matrices for NGS experiments: QC parameters for TSO500 included DNA integrity number >7 and RNA integrity number >7, size distribution of 200-500 bp for library construction and >90% on-target enrichment with uniform coverage. Under optimal storage conditions with minimal nucleic acid degradation, a ~90% success rate (108 out of 120 paired DNA/RNA samples) in sequencing with quality metrics was reported.
The Ion Torrent Oncomine Comprehensive Assay Panel v3 (OCP; cat. no. A35805; Thermo Fisher Scientific, Inc.) was used as the default CGP for the VGH-TAYLOR study, which enabled the detection of 161 cancer-related genes and the identification of single nucleotide variants (SNVs), CNVs, gene fusions and indels. A total of 10 ng of DNA and RNA sample input was required. Libraries were generated according to the standard protocols, which was constructed with the Ion AmpliSeq Library Kit Plus (cat. no. 4488990; Thermo Fisher Scientific Inc.), and were multiplexed for templating on the Ion OneTouch 2 System and subsequently sequenced on the Ion GeneStudio S5 Prime System (Thermo Fisher Scientific, Inc.) using the Ion 318 Chip Kit (cat. no. 488146; Thermo Fisher Scientific Inc.) with single-end semiconductor-based sequencing and a read length of 200 bp, according to the manufacturer's instructions (guide MAN0015885 Revision C of Thermo Fisher Scientific, Inc.). Sequencing data were analyzed, aligned and annotated through Torrent Suite v5.10.0 (Thermo Fisher Scientific, Inc.) and Ion Reporter v5.10 (Thermo Fisher Scientific, Inc.) software with the default Coverage Analysis (v5.10.0.3), Sample ID (v5.10.0.1) and Variant Caller (v5.10.0.18) plugin. Variants were further analyzed and interpreted for clinical actionability using the Oncomine Knowledgebase Reporter (Thermo Fisher Scientific, Inc.) database. The coverage metrics indicated that the number of mapped reads ranged from 4 to 6 million, with a mean depth of 1,100-1,600 times.
TSO500 (cat. no. 20032626; Illumina, Inc.) was designed to identify known and emerging tumor biomarkers, using both DNA and RNA from tumor samples. These pan-cancer biomarkers aligned with key guidelines and clinical trials (22,23), including 523 genes for assessment of DNA and RNA variant types, plus microsatellite instability, tumor mutational burden and homologous recombination deficiency (optional). Libraries were prepared according to the manufacturer's guidelines from up to 80 ng DNA and 40 ng of RNA, using the TSO 500 library prep kit (cat. no. 20028216; Illumina, Inc.). Adapter ligation with unique molecular identifiers (UMIs) was performed with target fragments amplified and indexed. NGS was performed with a NextSeq 2000 sequencing system optimized for a minimum read length of 2x101 bp, operated by the Department of Pathology and Laboratory Medicine of the Taipei Veterans General Hospital using the NextSeq 1000/2000 P2 Reagents (300 cycles) v3 (Illumina, Inc.). Data were analyzed with the TSO500 Local App v2.2 (Illumina, Inc.), and variant call format files were further processed and annotated with the PierianDx software version CGW_v6.20 (PierianDx), which offered an integrated interpretation. The read collapsing analysis step executed an algorithm that collapses sets of reads (known as families) with similar genomic locations into representative sequences using UMI tags. Median exon fragment coverage across all exon bases was ≥150.
The difference in sequencing technology between amplicon-based (OCP) and hybrid capture-based (TSO500) methods has implications for detecting genetic variants, especially in homopolymer regions, consisting of a series of consecutive identical bases (26). Hybridization capture-based approaches show better uniformity, which can be relevant for detecting variants in homopolymer regions. Amplicon-based methods are faster and cost-effective but prone to errors in homopolymer regions. Hybrid capture methods are more reliable for these regions due to uniform coverage and reduced PCR bias, but they are more resource-intensive. Final reports from the medium- (OCP) and large-sized (TSO500) panels and accompanied tab-separated values files were collected. Variants reported from actionable genes were the primary endpoints in the current study. Clinical actionability was defined by the joint consensus of the Association for Molecular Pathology, American Society of Clinical Oncology and College of American Pathologists, published in 2017(27). Additional annotations for actionability and OncoPrinter (28,29) visualization were conducted using the OncoKB database (30) and European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of molecular Targets (ESCAT) criteria (11,12). Clinical actionability was categorized as follows: i) Tier I, actionability indicated an alteration-drug match associated with improved outcome in clinical trials; ii) tier II, anti-tumor activity was associated with the matched alteration-drug but lacked prospective outcome data; and iii) tier III, the matched drug-alteration led to clinical benefit in another tumor type other than the tumor of interest. Polymorphisms were identified through population databases, such as the Single Nucleotide Polymorphism Database (31), the Genome Aggregation Database (32), 1000 Genomes Project and Exome Aggregation Consortium (33). Variants with minor allele frequency >1% were excluded. Variants outside the targeted regions of the sequencing panel were masked. Considering the sequencing depth, a 10% variant allele frequency (VAF) threshold assumed standard sequencing depths (500-1,000 times). The limit of detection was set to 5% for SNVs/indels and VAF #x003C;10% was considered a low-VAF status. An average copy number ≥4 was interpreted as a gain (amplification) and #x003C;1 as a loss (deletion). The concordance of filtered actionable genes (AKT1, BRCA1/2, PALB2, ERBB2, PIK3CA and PTEN) and TP53 based on the original reports between the two panels was calculated (concordant variants divided by the sum of both concordant and discordant variants) and reported.
ESCAT-defined actionable genes, including fusions, amplifications, copy number gains, point mutations and indels, were extracted for downstream analysis. Categorical variables are presented as numbers and percentages. Official reports from the Ion Reporter and the Oncomine Knowledgebase Reporter, as well as those from Pierian Dx software were released to all participants when requested. In addition, primary care physicians received the same reports once they were available, augmenting clinical decision making.
A total of 108 patients with breast cancer from the VGH-TAYLOR study with adequate remaining nucleic acid (both DNA and RNA) were recalled. After explaining the purpose of this re-sequencing study, all participants signed informed consent forms and their specimens were assayed with the large-sized CGP, TSO500 panel. There were 54 patients in group 1A, 6 in group 1B, 25 in group 2, 5 in group 3-I, 7 in group 3-II and 11 biobank/retrospective cohort breast cancer samples. Early-stage breast cancer (groups IA and 2) constituted the majority of samples (73.1%, n=79). Four patients initially classified as TNBC at the time of enrollment were re-tested by FISH and found to be HER2-positive. Due to discrepancy in the interrogated genes (523 vs. 161 genes), only actionable genes listed by the ESCAT criteria for breast cancer were analyzed, as follows: i) Tier IA: ERBB2 amplification, BRCA1/2 germline mutation and PIK3CA mutation; ii) tier IC: NTRK translocation; iii) tier IIA: PTEN loss and ESR1 mutation; iv) tier IIB: AKT1 mutation and ERBB2 mutation; v) tier IIIA: BRCA1/2 somatic mutation and MDM2 amplification; and vi) Tier IIIB: ERBB3 mutation (11,12).
Fig. 1A shows the mutational landscape of ESCAT-defined actionable genes among Taiwanese patients with TNBC examined with TSO500. Among 108 Taiwanese patients with breast cancer (all samples were TNBC except for four samples that were HER2-positive), PIK3CA was the most common actionable gene (39%, n=42), including fusion, amplification, copy number gain and point mutation, followed by BRCA2, including fusion, copy number gain, heterozygous loss, truncating and point mutation (24%, n=26). BRCA1 variants, including fusions, truncating and point mutations were detected in 12% (n=13) of samples. ERBB2 amplification, copy number gain, in-frame and point mutations were identified in 13% (n=14) of the study population. Importantly, among three breast cancer samples with HER2 amplification reported by TSO500, two coincided with the four clinically HER2-positive (overexpression) cases.
PTEN mutations including heterozygous/homozygous loss and truncating/in-frame/point mutations were reported in 15% (n=16) of breast cancer samples. Copy number gain and point mutations in ERBB3 were identified in 10% (n=11) of the samples, while ESR1 (2.8%, n=3; copy number gain/heterozygous loss and point mutation) and MDM2 variants (1.9%, n=2; amplification and heterozygous loss) were infrequently mutated in the Taiwanese population. No NTRK translocations were reported. Table SI presents the actionable mutations assayed with TSO500 CGP.
Fig. 1B shows the mutational landscape of actionable genes reported by OCP. Compared with TSO500, OCP reported a higher number of ERBB2 variants (19 vs. 13%), most of which came from higher proportion of missense mutations with unknown significance. Of the four clinical HER2-positive breast cancer samples, none reported a HER2 alteration, except for another HER2-equivocal case associated with an ERBB2 I665V missense mutation, which was a variant of uncertain significance (VUS). Conversely, the frequencies of BRCA1 and BRCA2 variants were lower than those reported from TSO500 (BRCA1, 6 vs. 12%; and BRCA2, 5 vs. 24%). PIK3CA variants and PTEN mutations were also less frequent than those with TSO500 (28 vs. 39% and 6 vs. 15%, respectively). No alterations were identified by OCP in ESR1, MDM2 and ERBB3. Table SII details the actionable mutations identified with OCP.
For benchmark comparisons, the four HER2-positive samples were excluded, and the analysis was focused on tumor DNA sequence variants from actionable genes. Amino acid change (protein coordinate), genomic and transcript-dependent cDNA coordinates were used to identify variants reported as least once from either TSO500, OCP or both (Table I). Among 272 variants, 94 (34.6%) were identified by both platforms; therefore, the concordance rate was slightly higher than one-third based on the original reports. To understand the mechanisms underpinning the high discordance between TSO500 and OCP, a manual review of conflicting variants was conducted with all binary alignment map (BAM) files visualized through integrative genomic viewer by an author who is expert in precision oncology and bioinformatics (34). Fig. S1 provides such an example.
Fig. 2 shows the interpretation categories of the comparison: A total of 25 (9.2%) variants were the same with different annotations, 34 (12.5%) were beyond the OCP targeted regions, 21 (7.7%) were benign polymorphisms called by OCP but filtered out by TSO500, 15 (5.5%) were discordant variants with low variant allele frequency (VAF#x003C;10%) while TSO500 were more accurate in 83 cases (30.5%). After discarding out-of-targeted region variants, benign and low-VAF variants, the concordance rate approached 60% (119 of 202 variants, 58.9%) and the large-sized panel (TSO500) detected more variants even for the same set of actionable genes and TP53. It deserves notice that certain variants were called by OCP but ignored by TSO500, which were proven to be homopolymer regions, including variants in BRCA1 (n=2), BRCA2 (n=2), PALB2 (n=1) and TP53 (n=3), as well as a misalignment with PTEN in one case and with TP53 in two cases (data not shown).
Fig. 3A and B and Table II present concordant and discordant variants among actionable genes between two CGP panels. AKT1 E17K exhibited a perfect concordance (100%, n=3), followed by PIK3CA (78%, n=29) and TP53 (52%, n=49). With manual corrections of polymorphism and aliasing variants, the concordance of TP53 (71%, n=67), ERBB2 (69%, n=20), PTEN (55%, n=6), BRCA1, BRCA2 and PALB2 (29, 17 and 8%, respectively) was enhanced substantially. Most of the discordance came from those detected by TSO500 only, including variants in BRCA1 (65%, n=11), BRCA2 (83%, n=29) and PALB2 (75%, n=9). Some variants were undetectable by OCP due to out-of-target regions or low VAF, which occurred in all ERBB3 and ESR1 variants, as well as NTRK1 and NTRK3 (76 and 71%, respectively) variants. True conflicting calling results were rare, including 3 TP53 (3%), 2 PIK3CA (5%) and 1 ERBB2 (3%) variants.
Table IIDeconvolution of variants among actionable genes and TP53 with concordant (original report and manual inspection) and discordant (outside targeted region/low VAF), conflicting results and TSO500-detected only results. |
To achieve the goal of personalized and precision medicine, CGP has been recognized as a key tool that can potentially transform cancer risk prediction, detection, diagnosis, treatment and monitoring. CGP can augment clinical decision-making in the form of either in vitro diagnostics (IVD) or companion diagnostics. Most importantly, the continuous price reduction across small- to large-sized CGP panels has improved patient access (35). Consequently, therapy assignments for newly diagnosed breast cancers and monitoring of patients who are in treatment are expected to benefit from CGP.
In the context of CGP, actionable genes refer to specific genetic alterations or mutations in a patient's (tumor) DNA that can guide therapeutic decisions or clinical interventions. These genetic changes are considered ‘actionable’ because they suggest potential treatments or clinical strategies that can directly impact the patient's care. This may include targeting specific mutations with precision medicine therapies, selecting clinical trials based on the genomic profile, such as basket or umbrella trials, or monitoring the patient for disease progression. Actionable genes typically include those that are associated with known therapies (such as targeted therapies and immunotherapies, including PARP inhibition for pathogenic BRCA1/2 mutations), have established clinical guidelines for intervention based on their presence, or may offer prognostic or predictive value for disease outcomes, guiding treatment options (1-5). In the present study, the performance of medium- to large-sized gene panels in a TNBC patient cohort was compared. Both OCP and TSO500 are used worldwide, but there are few direct comparison studies for breast cancer actionable genes in the literature (18,19,36,37).
The four HER2-positive breast cancer cases provided an opportunity to evaluate the association between clinical HER2-status and NGS-based ERBB2 CNVs. None of these cases were reported as HER2-amplified by OCP while two of three HER2-amplified cases reported by TSO500 coincided with the four clinically HER2-positive breast cancers. A higher number of HER2-gained breast cancers were identified by TSO500, indicating the potential for anti-HER2 targeting therapy. As NTRK translocation was not listed as the targeted alteration of OCP, the detectability of structure variants (CNV and fusion) was not compared further.
In the present study, the genes and variants reported from either OCP v3 or TSO500 when genomic, cDNA and protein coordinates were used for variant sorting were detailed. This subset represented the most actionable variants revealed by medium- and large-sized CGP. AKT1 is an intra-cellular kinase and is predominantly altered in breast and endometrial cancer. The AKT1 E17K is the most common alteration across various tumor types, including breast and gynecological cancers (38,39). The CAPItello-291 phase III trial demonstrated that capivasertib (AZD5363) and fulvestrant combination therapy resulted in markedly longer progression-free survival compared with the placebo and fulvestrant groups (40). The three AKT1 E17K-mutant cases in the present study were identified by both panels.
There were two BRCA1 (S405* and R1203*) and one BRCA2 (S521*) truncating mutations, and reflex germline testing should precede if PARP inhibitor was considered for these patients, as only germline BRCA1/2 mutations are actionable in breast cancer (41). The BRCA1 c.5470-1G>A SNP has been recognized as pathogenic by both the Breast Cancer Information Core and Consortium of Investigators of Modifiers of BRCA1/2 (42,43). The BRCA2 S521* truncating mutation impairs nuclear localization of BRCA2, which is essential for normal BRCA2 function (44). Despite common truncating and frameshift mutations (five for BRCA1 and three for BRCA2) called by both panels, only TSO500 revealed more suspicious variants (eight variants among 9 cases for BRCA1 and 13 variants among 21 subjects), while OCP was flawed by spurious mutations (BRCA1: S1180fs and T1376fs; and BRCA2: S538fs, T598fs, S973fs, E33*, T912fs, S3041fs and S3147fs).
PALB2 K353fs is a truncating mutation in a tumor suppressor gene and therefore is likely oncogenic. The phase II TBCRC 048 study also indicated the predictive power of the germline PALB2 mutation for metastatic breast cancer, as a high response rate (82%) was observed when olaparib, a PARP inhibitor, was used (45). In the present study, TSO500 identified three additional variants in 5 cases while OCP reported 1 false positive case with up to four variants (D1125fs, N368fs, S357fs and N342fs). Tumor-only sequencing made differentiating germline from somatic BRCA1/2 and PALB2 mutations challenging. However, LOH-germline inference calculator and the somatic-germline-zygosity algorithms helped distinguish these mutations, with one somatic and eight germline BRCA1/2 mutations identified (46).
One patient harbored two mutations in ERBB2 (L811V and L839R) and another with the L725S mutation was recognized by both platforms, none of which were among the oncogenic mutations approved by U.S. Food and Drug Administration for the use of neratinib, a pan-HER kinase inhibitor that binds irreversibly to the ATP-kinase domain of HER2 inhibiting the downstream phosphorylation of AKT and MAPK (47). TSO500 identified three additional ERBB2 mutations (P1140A, V743_M744insHV and Y742_A745dup). It is important to note that TSO500 detected more complex mutations (such as Y742_A745dup and V743_M744insHV, both classified as VUS) than did OCP.
Certain PIK3CA hotspot mutations have been identified from the SOLAR-1 trial for the usage of alpelisib, a selective PI3K-α inhibitor, namely C420R, E542K, E545A, E545D, E545G, E545K, Q546E, Q546R, H1047L, H1047R and H1047Y (48). Our previous study also observed double mutations of PIK3CA among the Taiwanese population (21). Only four cases with E726K, G1049R, N345I and Q546R mutations were outside the hotspot region, while two cases with D350N or D725N were co-mutant with other hotspots, which were only reported by TSO500. Consequently, both platforms identified the same number of PIK3CA-altered breast cancer cases.
Conversely, selective PI3K-β inhibitors, GSK2636771 and AZD8186, are ATP competitors and have shown preclinical antitumor response and durability for PTEN-deficient solid tumors including TNBC (49). The PTEN c.1026+1G>A SNP has been reported as clinically pathogenic at least twice (Invitae Clinical Genomics Group and ClinGen PTEN Variant Curation Expert Panel), with germline origin (50,51). The remaining five consensual variants (E150*, E43fs, P38fs, R130* and Q245*) were truncating mutations; TSO500 identified two additional cases with PTEN mutations (G127_G129del and V290Sfs*8) and OCP called one inappropriate variant due to misalignment (c.386G>A).
Quite a few TP53 alterations were identified by both OCP and TSO500. Despite not being currently druggable, TP53 may act as an independent prognostic factor for early and advanced-stage cancer with a wide range of mutational frequency (52). It has been argued that all TP53 mutations from tumor-only sequencing are somatic, and rarely representative of the germline Li-Fraumeni syndrome (53). TSO500 identified more variants than OCP did, while the latter reported three cases with V73fs in homopolymer region and one case with misaligned V73fs mutation, both of which were spurious.
A total of 272 variants were identified among actionable genes and TP53 from at least one panel. Interestingly, the concordance rate from original reports was low at slightly more than one-third (34.6%). With extensive bioinformatics analyses and manual curation, three-fifths of concordance (58.9%) were achieved after exclusion of unfiltered polymorphisms, low-VAF variants and those beyond the targeted scope of OCP. Among inconsistent variants, some were identical after manual inspections, further indicating the necessity of analytical abilities and domain knowledge in genomic nomenclature (54). Furthermore, an additional 62 variants were revealed by TSO500, indicating missed opportunities by OCP, which may be explained by fundamental discrepancies in sequencing technology. The accuracy of the amplicon-based OCP vs. the hybrid capture-based TSO500 panel can be influenced by their underlying methodologies. Amplicon-based methods utilize PCR amplification with sequence-specific primers to enrich target regions. By contrast, hybrid capture sequencing employs hybridization-based capture to enrich for desired genomic regions. Hybrid capture may offer a more robust and accurate approach, especially for clinical applications (55).
In the context of TNBC research, a VAF cut-off of #x003C;10% is used to filter out low-confidence or likely spurious SNVs identified through NGS of tumor samples (56,57). The threshold may be beneficial to eliminate technical, background noise and subclonal mutations, as tumor samples can be contaminated with normal cells, and low-level DNA damage or degradation can introduce background noise that interferes with accurate SNV calling. In addition, tumors are often composed of multiple subclones with distinct genetic profiles. SNVs present in minor subclones may have VAFs #x003C;10%, making them difficult to distinguish from background noise (58). A VAF cut-off of 5-10% is commonly used in various cancer genomics studies, including those focused on TNBC. This range is considered to achieve a reasonable balance between sensitivity (detecting true SNVs) and specificity (excluding false positives) (59).
Despite being one of the pioneers to directly compare two commercialized targeted panels, there were some limitations to the present study. First, sequencing was not conducted concurrently, and the 1-2 year delay of the TSO500 analysis following OCP may have introduced some bias from nucleic acid degradation, despite all samples being stored under temperature-controlled conditions. Second, variants called by each panel were considered based on the standard or formal algorithm of each platform (Ion Reporter/Oncomine Knowledgebase Reporter for OCP and PierianDx for TSO500), which limited the comparability between distinct platforms. For unbiased comparison, the same aligner, caller and annotator should be applied. However, browser extensible data files were unavailable from manufacturers to confirm the jointly interrogated regions. On the other hand, all commercial CGP solutions are under regulation as either laboratory developed tests or IVD, and practically these pre-set algorithms should not be modified arbitrarily to enhance reproducibility. Despite this, the present study conducted an exhaustive bioinformatic analysis from BAM files to dissect the conflicting results from the same samples. Third, NTRK is not within the targeted fusion genes of OCP; therefore, a comparison with TSO500 was not possible. Moreover, NTRK sequence variants, amplifications or fusions are not actionable for breast cancer. As larotrectinib and entrectinib are approved in a number of countries, NTRK fusion as a tumor-agnostic marker is important, and its detection should be incorporated into CGP for breast cancer (60). Fourth, as most cases are triple negative phenotypically, a cohort of ~100 patients with TNBC is suitable for a comparative sequencing panel study. However, if the goal is to perform more detailed subgroup analyses or identify rare mutations, a larger cohort may be more appropriate. In the future, conducting a prospective and parallel study across distinct CGP platforms could reveal whether larger sequencing panels justify their higher costs by improving the detection of actionable mutations, refining novel therapeutic strategies, and characterizing the complex genomic landscape of TNBC. Furthermore, it would provide critical insights to optimize genomic profiling for other breast cancer subtypes, advancing both research and clinical care.
As an observational study, the present study could not directly assess the impact of CGP on treatment outcomes, as no intervention was performed. During the study period, the compassionate use of alpelisib (a selective PI3K inhibitor) benefited nine patients, and the out-of-pocket use of olaparib (a PARP inhibitor) benefited two patients, demonstrating the actionability of the testing. Since May 2024, NGS, specifically whole-exome sequencing for BRCA1/2, has been reimbursed in Taiwan for patients with stage II or higher TNBC. The Regulations of Special Medical Techniques of Taiwan require that all NGS testing results (including both whole-exome sequencing and targeted panels) be submitted to the National Health Research Institutes Biobank. It is anticipated that cost-effective implementation of larger NGS panels in routine clinical practice will be possible in the future, once sufficient data from NGS results and accompanying clinical outcomes are available.
In conclusion, the present study comparing medium- and large-sized sequencing panels in a cohort of patients with TNBC may provide critical insights into the balance between actionable gene findings, cost and complexity. TSO500, the larger panel, detected more variants than OCP did, even from the same set of ESCAT-defined actionable genes and TP53. Conversely, a proportion of inconsistent variants could be manually curated and were identical with aliasing coordinates or starting positions. Finally, there were variants detectable only by TSO500, indicating potential and fundamental differences in sequencing technology, bioinformatic algorithms and variant filtering. The value of a large-sized panel in clinical usage is ascertained, considering the beneficiaries of PARP inhibition for the higher number of BRCA1/2 and PALB2 mutations detected, as indicated by the National Comprehensive Cancer Network and ESMO guidelines (61,62). Given the experience from the present study, the updated Oncomine Comprehensive Assay Plus with >500 genes profiled is anticipated in future studies (63). The results could guide panel selection for precision oncology, ensuring optimal clinical outcomes while maximizing resource efficiency. This approach would be particularly impactful in refining strategies for TNBC management.
The authors would like to thank Dr Morris Chang (retired from the Taiwan Semiconductor Manufacturing Company) for professional consultation about the technical feasibility of the present study. The authors also appreciate the kind support with NGS experiments from the Illumina team (Illumina, Inc.; San Diego, CA), including Dr Mark M. Wang, Dr Vincent Hsieh, Mr. Thomas Tang, Mrs. Jora Lin, Mr. Dr Wang and Mr. Jerry Cheng throughout the execution of this study.
Funding: This work was supported in part by Taipei Veterans General Hospital research fund grant nos. V110E-005-3, V111E-006-3, V112E-004-3 and V112C-013, Melissa Lee Cancer Foundation (grant no. MLCF_V114_B11404) and National Science and Technology Council (grant no. NSTC 111-2314-B-075-063-MY3).
The data generated in the present study may be found in the Sequence Read Archive database under accession number PRJNA1256731 or at the following URL: https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1256731.
CCH and LMT conceived the study. CCH drafted the manuscript. YCY conducted bioinformatics analyses. YFT, YSL, TCC and CYL collected samples and certified the NGS reports. HLH conducted NGS experiments. CCH and YCY confirm the authenticity of all the raw data. LMT approved the final submission. All authors read and approved the final manuscript.
The study protocol was reviewed and approved by the Institutional Review Board of Taipei Veterans General Hospital (approval nos. 2021-01-007B and 2023-08-002B; Taipei, Taiwan). Participants provided written informed consent for both the prospective/retrospective VGH-TAYLOR study (approval no. 2021-01-007B) and for the retrospective re-sequencing study (approval no. 2023-08-002B).
All participants agreed to the publication of the present study, with all identifying information removed.
The authors declare that they have no competing interests.
|
Huang CC, Yeh YC, Ho HL, Liu CY, Tsai YF and Tseng LM: Comprehensive genomic profiling of Taiwanese patients with breast cancer using a novel targeted panel: Preliminary analyses from a prospective triple-negative cohort. J Clin Oncol. 41 (Suppl 16)(e12553)2023. | |
|
Wang L, Zhai Q, Lu Q, Lee K, Zheng Q, Hong R and Wang S: Clinical genomic profiling to identify actionable alterations for very early relapsed triple-negative breast cancer patients in the Chinese population. Ann Med. 53:1358–1369. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Tierno D, Grassi G, Scomersi S, Bortul M, Generali D, Zanconati F and Scaggiante B: Next-generation sequencing and triple-negative breast cancer: Insights and applications. Int J Mol Sci. 24(9688)2023.PubMed/NCBI View Article : Google Scholar | |
|
O'Haire S, Degeling K, Franchini F, Tran B, Luen SJ, Gaff C, Smith K, Fox S, Desai J and IJzerman M: Comparing survival outcomes for advanced cancer patients who received complex genomic profiling using a synthetic control arm. Target Oncol. 17:539–548. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Cifuentes C, Lombana M, Vargas H, Laguado P, Ruiz-Patiño A, Rojas L, Navarro U, Vargas C, Ricaurte L, Arrieta O, et al: Application of comprehensive genomic profiling-based next-generation sequencing assay to improve cancer care in a developing country. Cancer Control. 30(10732748231175256)2023.PubMed/NCBI View Article : Google Scholar | |
|
Almansour NM: Triple-negative breast cancer: A brief review about epidemiology, risk factors, signaling pathways, treatment and role of artificial intelligence. Front Mol Biosci. 9(836417)2022.PubMed/NCBI View Article : Google Scholar | |
|
Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P and Narod SA: Triple-negative breast cancer: Clinical features and patterns of recurrence. Clin Cancer Res. 13:4429–4434. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Foulkes WD, Smith IE and Reis-Filho JS: Triple-negative breast cancer. N Engl J Med. 363:1938–1948. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Stewart RL, Updike KL, Factor RE, Henry NL, Boucher KM, Bernard PS and Varley KE: A multigene assay determines risk of recurrence in patients with triple-negative breast cancer. Cancer Res. 79:3466–3478. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Jie H, Ma W and Huang C: Diagnosis, prognosis, and treatment of triple-negative breast cancer: A review. Breast Cancer (Dove Med Press). 17:265–274. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Mateo J, Chakravarty D, Dienstmann R, Jezdic S, Gonzalez-Perez A, Lopez-Bigas N, Ng CKY, Bedard PL, Tortora G, Douillard JY, et al: A framework to rank genomic alterations as targets for cancer precision medicine: The ESMO scale for clinical actionability of molecular targets (ESCAT). Ann Oncol. 29:1895–1902. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Condorelli R, Mosele F, Verret B, Bachelot T, Bedard PL, Cortes J, Hyman DM, Juric D, Krop I, Bieche I, et al: Genomic alterations in breast cancer: Level of evidence for actionability according to ESMO scale for clinical actionability of molecular targets (ESCAT). Ann Oncol. 30:365–373. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Bianchini G, Balko JM, Mayer IA, Sanders ME and Gianni L: Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 13:674–690. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Masci D, Naro C, Puxeddu M, Urbani A, Sette C, La Regina G and Silvestri R: Recent advances in drug discovery for triple-negative breast cancer treatment. Molecules. 28(7513)2023.PubMed/NCBI View Article : Google Scholar | |
|
Baden J, Zhao C, Pratt J, Kirov S, Pant A, Seminara A, Green G, Bilke S, Deras I, Fabrizio DA and Pawlowski T: 90PD-Comparison of platforms for determining tumour mutational burden (TMB) in patients with non-small cell lung cancer (NSCLC). Ann Oncol. 30 (Suppl 5)(v25)2019. | |
|
Wei B, Kang J, Kibukawa M, Arreaza G, Maguire M, Chen L, Qiu P, Lang L, Aurora-Garg D, Cristescu R and Levitan D: Evaluation of the TruSight oncology 500 assay for routine clinical testing of tumor mutational burden and clinical utility for predicting response to pembrolizumab. J Mol Diagn. 24:600–608. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Liu CY, Huang CC, Tsai YF, Chao TC, Lien PJ, Lin YS, Feng CJ, Chen JL, Chen YJ, Chiu JH, et al: VGH-TAYLOR: Comprehensive precision medicine study protocol on the heterogeneity of Taiwanese breast cancer patients. Future Oncol: Oct 19, 2021 (Epub ahead of print). | |
|
Huang CC, Tsai YF, Liu CY, Chao TC, Lien PJ, Lin YS, Feng CJ, Chiu JH, Hsu CY and Tseng LM: Comprehensive molecular profiling of Taiwanese breast cancers revealed potential therapeutic targets: prevalence of actionable mutations among 380 targeted sequencing analyses. BMC Cancer. 21(199)2021.PubMed/NCBI View Article : Google Scholar | |
|
Huang CC, Tsai YF, Liu CY, Lien PJ, Lin YS, Chao TC, Feng CJ, Chen YJ, Lai JI, Phan NN, et al: Prevalence of tumor genomic alterations in homologous recombination repair genes among taiwanese breast cancers. Ann Surg Oncol. 29:3578–3590. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Huang CC, Tsai YF, Liu CY, Lien PJ, Lin YS, Chao TC, Feng CJ, Chen YJ, Lai JI, Cheng HF, et al: Concordance of targeted sequencing from circulating tumor DNA and paired tumor tissue for early breast cancer. Cancers (Basel). 15(4475)2023.PubMed/NCBI View Article : Google Scholar | |
|
Chao TC, Tsai YF, Liu CY, Lien PJ, Lin YS, Feng CJ, Chen YJ, Lai JI, Hsu CY, Lynn JJ, et al: Prevalence of PIK3CA mutations in Taiwanese patients with breast cancer: A retrospective next-generation sequencing database analysis. Front Oncol. 13(1192946)2023.PubMed/NCBI View Article : Google Scholar | |
|
Gradishar WJ, Moran MS, Abraham J, Abramson V, Aft R, Agnese D, Allison KH, Anderson B, Burstein HJ, Chew H, et al: NCCN guidelines® insights: Breast cancer, version 4.2023. J Natl Compr Canc Netw. 21:594–608. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Gennari A, André F, Barrios CH, Cortés J, de Azambuja E, DeMichele A, Dent R, Fenlon D, Gligorov J, Hurvitz SA, et al: ESMO clinical practice guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol. 32:1475–1495. 2021.PubMed/NCBI View Article : Google Scholar | |
|
No authors listed. Pathologists' guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Breast Care (Basel). 5:185–187. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Wolff AC, Somerfield MR, Dowsett M, Hammond MEH, Hayes DF, McShane LM, Saphner TJ, Spears PA and Allison KH: Human epidermal growth factor receptor 2 testing in breast cancer: ASCO-college of american pathologists guideline update. J Clin Oncol. 41:3867–3872. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Schirmer M, Ijaz UZ, D'Amore R, Hall N, Sloan WT and Quince C: Insight into biases and sequencing errors for amplicon sequencing with the Illumina MiSeq platform. Nucleic Acids Res. 43(e37)2015.PubMed/NCBI View Article : Google Scholar | |
|
Li MM, Datto M, Duncavage EJ, Kulkarni S, Lindeman NI, Roy S, Tsimberidou AM, Vnencak-Jones CL, Wolff DJ, Younes A and 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. 19:4–23. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6(pl1)2013.PubMed/NCBI View Article : Google Scholar | |
|
Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, Rudolph JE, Yaeger R, Soumerai T, Nissan MH, et al: OncoKB: A precision oncology knowledge base. JCO Precis Oncol. 2017(PO.17.00011)2017.PubMed/NCBI View Article : Google Scholar | |
|
Phan L, Zhang H, Wang Q, Villamarin R, Hefferon T, Ramanathan A and Kattman B: The evolution of dbSNP: 25 Years of impact in genomic research. Nucleic Acids Res. 53:D925–D931. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Chen S, Francioli LC, Goodrich JK, Collins RL, Kanai M, Wang Q, Alföldi J, Watts NA, Vittal C, Gauthier LD, et al: A genomic mutational constraint map using variation in 76,156 human genomes. Nature. 625:92–100. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Karczewski KJ, Weisburd B, Thomas B, Solomonson M, Ruderfer DM, Kavanagh D, Hamamsy T, Lek M, Samocha KE, Cummings BB, et al: The ExAC browser: Displaying reference data information from over 60 000 exomes. Nucleic Acids Res. 45:D840–D845. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Robinson JT, Thorvaldsdottir H, Turner D and Mesirov JP: igv.js: An embeddable JavaScript implementation of the Integrative Genomics Viewer (IGV). Bioinformatics. 39(btac830)2023.PubMed/NCBI View Article : Google Scholar | |
|
Wolff L and Kiesewetter B: Applicability of ESMO-MCBS and ESCAT for molecular tumor boards. Memo. 15:190–195. 2022. | |
|
Huang CC, Yeh YC, Cheng HF, Chen BF, Liu CY, Tsai YF, Ho HL and Tseng LM: Comprehensive genomic profiling of Taiwanese triple-negative breast cancer with a large targeted sequencing panel. J Chin Med Assoc: Jun 20, 2025 (Epub ahead of print). | |
|
Loderer D, Hornáková A, Tobiášová K, Lešková K, Halašová E, Danková Z, Biringer K, Kúdela E, Rokos T, Dzian A, et al: Comparison of next-generation sequencing quality metrics and concordance in the detection of cancer-specific molecular alterations between formalin-fixed paraffin-embedded and fresh-frozen samples in comprehensive genomic profiling with the Illumina® TruSight oncology 500 assay. Exp Ther Med. 29(64)2025. | |
|
Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, Robbins CM, Hostetter G, Boguslawski S, Moses TY, Savage S, et al: A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature. 448:439–444. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Kalinsky K, Hong F, McCourt CK, Sachdev JC, Mitchell EP, Zwiebel JA, Doyle LA, McShane LM, Li S, Gray RJ, et al: Effect of capivasertib in patients with an AKT1 E17K-mutated tumor: NCI-MATCH subprotocol EAY131-Y nonrandomized trial. JAMA Oncol. 7:271–278. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Turner NC, Oliveira M, Howell SJ, Dalenc F, Cortes J, Gomez Moreno HL, Hu X, Jhaveri K, Krivorotko P, Loibl S, et al: Capivasertib in hormone receptor-positive advanced breast cancer. N Engl J Med. 388:2058–2070. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Daly GR, AlRawashdeh MM, McGrath J, Dowling GP, Cox L, Naidoo S, Vareslija D, Hill ADK and Young L: PARP inhibitors in breast cancer: A short communication. Curr Oncol Rep. 26:103–113. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Szabo C, Masiello A, Ryan JF and Brody LC: The breast cancer information core: Database design, structure, and scope. Hum Mutat. 16:123–131. 2000.PubMed/NCBI View Article : Google Scholar | |
|
Chenevix-Trench G, Milne RL, Antoniou AC, Couch FJ, Easton DF and Goldgar DE: CIMBA. An international initiative to identify genetic modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: The consortium of investigators of modifiers of BRCA1 and BRCA2 (CIMBA). Breast Cancer Res. 9(104)2007.PubMed/NCBI View Article : Google Scholar | |
|
Spain BH, Larson CJ, Shihabuddin LS, Gage FH and Verma IM: Truncated BRCA2 is cytoplasmic: Implications for cancer-linked mutations. Proc Natl Acad Sci USA. 96:13920–13925. 1999.PubMed/NCBI View Article : Google Scholar | |
|
Tung NM, Robson ME, Ventz S, Santa-Maria CA, Nanda R, Marcom PK, Shah PD, Ballinger TJ, Yang ES, Vinayak S, et al: TBCRC 048: Phase II study of olaparib for metastatic breast cancer and mutations in homologous recombination-related genes. J Clin Oncol. 38:4274–4282. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Cheng HF, Tsai YF, Liu CY, Hsu CY, Lien PJ, Lin YS, Chao TC, Lai JI, Feng CJ, Chen YJ, et al: Prevalence of BRCA1, BRCA2, and PALB2 genomic alterations among 924 Taiwanese breast cancer assays with tumor-only targeted sequencing: Extended data analysis from the VGH-TAYLOR study. Breast Cancer Res. 25(152)2023.PubMed/NCBI View Article : Google Scholar | |
|
Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, Juric D, Quinn DI, Moreno V, Doger B, et al: HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature. 554:189–194. 2018.PubMed/NCBI View Article : Google Scholar | |
|
André F, Ciruelos EM, Juric D, Loibl S, Campone M, Mayer IA, Rubovszky G, Yamashita T, Kaufman B, Lu YS, et al: Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from SOLAR-1. Ann Oncol. 32:208–217. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Choudhury AD, Higano CS, de Bono JS, Cook N, Rathkopf DE, Wisinski KB, Martin-Liberal J, Linch M, Heath EI, Baird RD, et al: A phase I study investigating AZD8186, a potent and selective inhibitor of PI3Kβ/δ, in patients with advanced solid tumors. Clin Cancer Res. 28:2257–2269. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Chen HJ, Romigh T, Sesock K and Eng C: Characterization of cryptic splicing in germline PTEN intronic variants in Cowden syndrome. Hum Mutat. 38:1372–1377. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Mester JL, Ghosh R, Pesaran T, Huether R, Karam R, Hruska KS, Costa HA, Lachlan K, Ngeow J, Barnholtz-Sloan J, et al: Gene-specific criteria for PTEN variant curation: Recommendations from the ClinGen PTEN expert panel. Hum Mutat. 39:1581–1592. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Nykamp K, Anderson M, Powers M, Garcia J, Herrera B, Ho YY, Kobayashi Y, Patil N, Thusberg J, Westbrook M, et al: Sherloc: A comprehensive refinement of the ACMG-AMP variant classification criteria. Genet Med. 19:1105–1117. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Garrido-Navas MC, García-Díaz A, Molina-Vallejo MP, González-Martínez C, Alcaide Lucena M, Cañas-García I, Bayarri C, Delgado JR, González E, Lorente JA and Serrano MJ: The polemic diagnostic role of TP53 mutations in liquid biopsies from breast, colon and lung cancers. Cancers (Basel). 12(3343)2020.PubMed/NCBI View Article : Google Scholar | |
|
Zhou W, Chen T, Chong Z, Rohrdanz MA, Melott JM, Wakefield C, Zeng J, Weinstein JN, Meric-Bernstam F, Mills GB and Chen K: TransVar: A multilevel variant annotator for precision genomics. Nat Methods. 12:1002–1003. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Mosteiro M, Azuara D, Villatoro S, Alay A, Gausachs M, Varela M, Baixeras N, Pijuan L, Ajenjo-Bauza M, Lopez-Doriga A, et al: Molecular profiling and feasibility using a comprehensive hybrid capture panel on a consecutive series of non-small-cell lung cancer patients from a single centre. ESMO Open. 8(102197)2023.PubMed/NCBI View Article : Google Scholar | |
|
Fox EJ, Reid-Bayliss KS, Emond MJ and Loeb LA: Accuracy of Next generation sequencing platforms. Next Gener Seq Appl. 1(1000106)2014.PubMed/NCBI View Article : Google Scholar | |
|
Song P, Chen SX, Yan YH, Pinto A, Cheng LY, Dai P, Patel AA and Zhang DY: Selective multiplexed enrichment for the detection and quantitation of low-fraction DNA variants via low-depth sequencing. Nat Biomed Eng. 5:690–701. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Apostoli AJ and Ailles L: Clonal evolution and tumor-initiating cells: New dimensions in cancer patient treatment. Crit Rev Clin Lab Sci. 53:40–51. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Edsjö A, Gisselsson D, Staaf J, Holmquist L, Fioretos T, Cavelier L and Rosenquist R: Current and emerging sequencing-based tools for precision cancer medicine. Mol Aspects Med. 96(101250)2024.PubMed/NCBI View Article : Google Scholar | |
|
Cocco E, Scaltriti M and Drilon A: NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 15:731–747. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, Bonastre J, Castro E, Dienstmann R, Krämer A, et al: Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: A report from the ESMO precision medicine working group. Ann Oncol. 35:588–606. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Gradishar WJ, Moran MS, Abraham J, Abramson V, Aft R, Agnese D, Allison KH, Anderson B, Bailey J, Burstein HJ, et al: Breast cancer, version 3.2024, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 22:331–357. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Kang J, Na K, Kang H, Cho U, Kwon SY, Hwang S and Lee A: Prediction of homologous recombination deficiency from oncomine comprehensive assay plus correlating with SOPHiA DDM HRD solution. PLoS One. 19(e0298128)2024.PubMed/NCBI View Article : Google Scholar |