Abstract
Comprehensive genomic profiling (CGP) refers to the detailed genomic analysis of cancers for oncology patients. With the rapid development of next-generation sequencing (NGS) technologies, CGP has been widely applied to clinical practice and managing oncology patients. CGP can be performed on the tumor DNA and RNA, as well as non-tumor tissues (e.g., blood, pleural effusion, and ascites). In this article, we review the current evidence supporting the use of CGP in the management of oncology patients, both in real-world practice and the bridging to clinical trials. We also discuss the role of the molecular tumor board on the application of CGP in oncology patients. We provide an overview of the current scheme of CGP reimbursement in Taiwan and the precision oncology branch of the National Biobank Consortium of Taiwan. Finally, we discuss about the potential barriers and challenges of applying CGP in managing oncology patients and the future perspectives of CGP in precision oncology.
1. Background and introduction
Ever since the development of next-generation sequencing (NGS) and its growing application in scientific research and in clinical practice, the current scheme of managing oncology patients is experiencing a paradigm shift. NGS can provide in-depth analysis of various genomic alterations, including single nucleotide variations (SNVs), small insertions and deletions (indels), somatic copy number alterations, structural variations, and other genomic features [1]. In oncology patients, NGS is a powerful tool to inform genomic alterations, which may help the clinical management and potentially improve the treatment outcome. Comprehensive genomic profiling (CGP) refers to the detailed analysis of a patient's tumor at the genomic level. Compared to traditional molecular testing, NGS-based approaches profile a larger number of genes in a single test, providing a thorough overview of the genomic alterations of the cancer.
In precision oncology, substantial evidence supports the integration of CGP into routine care practice. CGP could identify predictive, prognostic, and diagnostic biomarkers in cancers, which may eventually tailor the management of oncology patients [2]. In this review article, we aim to review and summarize the growing evidence of integrating CGP in the management of oncology patients. We also discuss the role of the molecular tumor board (MTB) and summarize the current scheme of CGP reimbursement in Taiwan. Finally, we discuss some of the challenges and future direction in the application of CGP in precision oncology.
2. Overview of the NGS technologies
NGS is a massively sequencing technique that allows billions of sequencing processes to happen simultaneously in parallel on a solid surface (glass or beads) [3]. Short-read sequencing is the mainstream technology in NGS that generates sequences of less than 1000 bp (typically 150–300 bp) per read. Prior to the sequencing, library preparation is required to obtain templates corresponding to the target of interest for sequencing. Amplicon-based and hybridization capture-based methodologies are the two major approaches employed for target enrichment. Both methods are available in commercialized forms, but the performance may vary. For example, hybridization capture-based enrichment is more sensitive in detecting insertion and deletions (indels) of larger size [4]. On the other hand, amplicon-based enrichment is more sensitive in detecting variants with lower allele fractions [5]. Overall, both methods reach high concordance and accuracy in detecting small genetic variants [4].
Based on the region of genome enriched for sequencing, the NGS application can be categorized into three types: whole-genome sequencing (WGS), whole-exome sequencing (WES), and targeted-panel sequencing (TPS). WGS covers a complete genomic region, and WES captures more than 95 % of the coding regions. In contrast to WGS or WES, TPS often focuses on a panel of genes representing 2–3 % of coding regions [6]. These genes are often highly associated with clinical outcomes or responses to certain therapies. In addition, TPS reduces costs and data burden, but still offers greater sequencing depth and diagnostic power [6]. Currently, TPS is the most common method for CGP in clinical practice. Compared to targeted-panel sequencing, CGP that covers the complete coding mutations and gene fusions (i.e., WES and WGS) is a preferred method for predicting personalized tumor-associated neoantigen [7].
The number of genes included in a single CGP test ranges from 11 to 596 [8]. However, the most optimal size of gene panel covered in one CGP test remains unknown. Comparing to patients with advanced non-small cell lung cancer (NSCLC) who received CGP focusing a smaller gene panel (<53 genes), patients who received CGP with a larger gene panel had improved detection of actionable targets and greater use of matched therapies, both of which were associated with significant increases in survival [9]. However, the additional benefits of CGP with a larger size of gene panel for patients with other types of cancers remain unclear.
Despite the striking achievements of short-read sequencing in genomic profiling, this technology is still underpowered to resolve some genomic regions, including repetitive regions (e.g., centromere and telomere) and complex structural variations [10]. The emergence of long-read sequencing technologies, such as PacBio and Oxford Nanopore Technology, has potentiated the ability to tackle this problem by generating sequences up to tens of thousands or million bp per read [10]. Long-read NGS is also useful in phasing the somatic mutations and genotyping HLA molecules [11,12]. However, long-read NGS suffers from lower base-calling accuracy and much higher sequencing costs, which limits their application in precision oncology [13]. The evidence that supports the use of long-read NGS in clinical practice is scarce. Currently, the use of long-read NGS in precision oncology is primarily in the context of clinical research.
3. Genomic profiling using RNA as sequencing material
Gene fusions, which can be caused by intrachromosomal or interchromosomal rearrangement, may introduce fusion transcripts that translates to chimeric proteins, resulting in signaling activation and cancer promotion [14]. Oncogenic fusions can be found in 16.5 % of cancer cases and function as the sole driver in 1 % of them [14]. Currently, fusions involving ALK, ROS1, NTRK, RET, NRG1, MET, or FGFR genes are tier I clinical actionability across various cancer types [15]. Although these fusion variants can be detected with commercially available DNA-based assays [16], variants involving large introns, repetitive sequences, or unknown fusion partners may be overlooked in some circumstances. For detecting of these fusion variants, RNA-based assays are generally more sensitive than DNA-based assays [17,18].
RNA-based assays are the best method of choice to study variants with alternative splicing, some of which are oncogenic and susceptible to targeted therapies. For example, MET exon-14-skipping mutation is an uncommon genomic alteration in NSCLC that leads to skipping loss of exon 14 and converts to sensitivity to MET-targeted therapies [19]. In NSCLCs harboring MET exon-14-skipping variants, a study found that RNA-based assays are more sensitive than DNA-based assays [20].
However, RNA-based CGP requires high quality of RNA contained in the tissue, which can be suboptimal in some clinical samples. Currently, RNA-based genomic profiling assays are available for clinical use [21]. Overall, RNA-based genomic profiling is more sensitive in detecting gene fusions and alternative splicing variants. This approach may be critical for managing patients with malignancies that harboring these genomic alterations [22]. Table 1 summarizes the features and applications of different NGS assays using DNA or RNA as sequencing material.
Table 1.
Clinical applications of different NGS assays using DNA or RNA as sequencing material.
| Assay type | DNA-based | DNA-based | DNA-based | RNA-based |
|---|---|---|---|---|
| NGS method | Whole-genome sequencing | Whole-exome sequencing | Targeted-panel sequencing | RNA sequencing |
| Region of human genome covered | >99 % | ∼2 % | ∼0.05 % | 0.01–2 % |
| Number of coding genes covered | ∼20,000 | ∼20,000 | 50–1000 | 20–20,000 (Depend on the assay) |
| Average read depth per target region | 30–60 × | 50–200 × | 500–1000 × | (Depend on the transcript abundance) |
| Accuracy in detecting small variants (SNVs and indels) | Good | Good | Excellent | Poor |
| Accuracy in detecting copy number variation | Good | Intermediate | Poor | Poor |
| Accuracy in detecting noncoding variants | Excellent | Poor | Poor | Poor |
| Accuracy in detecting gene translocation | Good | Poor | (Probe-dependent) | Intermediate |
| Accuracy in detecting large structural variation | Good | Intermediate | Poor | Poor |
| Accuracy in detecting alternative splicing variants or splice site variants | Good | Intermediate | Poor | Excellent |
| Sequencing cost | $$$$ | $$ | $ | $$ |
4. CGP using non-tissue samples
Although tissue-based CGP remains the standard approach to identify actionable genomic aberrations, recent advancement has enabled CGP to be performed on non-tissue samples, such as peripheral blood, pleural effusion, or ascites [23]. Plasma circulating tumor DNA (ctDNA) has been found to represent the genomic alterations of primary tumors, highlighting the feasibility of CGP using ctDNA in clinical practice [24,25]. Published studies showed that the concordance of CGP between ctDNA and tissue samples was highly variable, ranging from 8.3 % to 93 % across different cancer types [23]. Despite the inconsistencies in recapitulating the genomic landscapes, the detection of oncogenic driver variants using cfDNA-based CGP demonstrated great sensitivity, ranging between 75 % and 93 % [23]. Clinical activity of targeted therapies or immunotherapy in patients with advanced NSCLC based on the CGP using ctDNA is currently under investigation [26,27].
Currently, CGP using ctDNA still faces many technical and biological challenges that limit its application in the management of oncology patients. The ctDNA in blood is often fragmented (with an average size of 150–300 bp) and low in concentration (5–10 pg/μL) [28]. This makes the detection of target variants extremely noisy and unstable. Overall, ctDNA detection rate was higher for clonal mutations (75–90 %) than subclonal mutations (20–30 %) [29]. In addition, some cancers are less prone to releasing ctDNA into the circulation, also known as the non-shedding tumors [30]. These include brain, kidney, thyroid, breast, and liver cancers [31]. Studies also reported a lower detection rate of ctDNA in colorectal cancer patients without liver metastasis compared to those with liver metastases [32]. These biological features may as well impact the sensitivity and the application of ctDNA testing in clinical practice.
Malignant pleural effusions and ascites are common complications in patients with advanced cancer. When tumor tissues are not available, these samples can be alternative options for molecular testing or CGP. When applied in malignant pleural effusions, CGP have comparable (65–87 %) concordance of molecular alteration with corresponding tissue biopsies [33,34]. The concordance was highly variant for malignant ascites (25–90 %) [35,36]. Although NGS can be applied to pleural effusion and ascites, more studies are needed to validate the accuracy of detecting clinical actionabilities in cancer patients. Nevertheless, CGP using non-tissue samples is still a valuable diagnostic tool in managing oncology patients.
5. The development and application of CGP in precision oncology
Underpinning precision oncology is the core concept of somatic mutations as the foundation of cancer development [37]. Many tumor-specific and tumor-agnostic mutations are well-proven to be predictive biomarkers of response or resistance to targeted therapies, with novel biomarkers continuously emerging [38]. Variants of non-actionable genes may inform prognosis and predict response to treatment. Conventional tests, such as immunohistochemistry staining, fluorescence in situ hybridization, Sanger sequencing, and quantitative polymerase chain reaction (qPCR), are reliable tools to identify these alterations in daily practice [39]. However, as the number of clinically meaningful biomarkers expands, CGP has greater efficiency and a shorter turn-around time compared to those of traditional testing [40]. Using NSCLC as an example, there are currently 11 genes have been approved as tier I actionable targets [41]. This includes oncogenic mutations in EGFR, ALK, ROS1, KRAS, MET, ERBB2, BRAF, NTRK, and RET genes. To date, there are 53 genes approved as level 1 actionable targets across all cancer types [41]. Some of the tumor-agnostic genomic alterations are rare but yet highly responsive to targeted therapies, such as BRAFV600E mutations, RET fusions, and NTRK fusions. CGP assay is also frequently used for the diagnosis of complex biomarkers, such as microsatellite instability (MSI), tumor mutational burden (TMB), and homologous recombination deficiency (HRD) [[42], [43], [44]]. Considering the number actionable genes are still expanding, CGP is an ideal diagnostic tool in clinical practice [45].
To evaluate whether CGP should be integrated in managing patients with advanced cancer, several large cohorts were conducted to address this issue. The SHIVA study that included 741 refractory cancer patients found that 40 % of patients had at least 1 druggable molecular alteration [46]. The MOSCATO-01 study that included 1035 patients with advanced cancer demonstrated that 49 % (411 of 843) of adult patients and 61 % (42 of 69) of pediatric patients had actionable molecular alterations [47,48]. The ProfiLER study included 1980 patients with refractory cancers who successfully underwent CGP and showed that 52 % of patients had at least 1 actionable mutation [49]. The Long March Pathway trial that included 520 Chinese cancer patients showed that 22 % of patients had actionable targets [50]. Of note, the frequencies of tumors harboring actionable targets vary across cancer types, with the highest in gastrointestinal stromal tumors, non-small cell lung cancer, and thyroid cancer [51]. In addition, the definition of clinical actionability has expanded substantially in the past few years. An overall increase from 8.9 % to 31.6 % in the fraction of tumors harboring a standard care (level 1 or 2) was observed in a large retrospective study [51]. Although progress has been made in expanding the use of CGP and recognizing clinical actionability, a continued unmet need for innovative therapeutic strategies remains a major issue in precision oncology, particularly for cancers with currently undruggable oncogenic drivers.
6. Evidence supporting the use of CGP in the management of oncology patients
The introduction of CGP allowed the cancer genome to be systematically studied, and guide the therapy in managing oncology patients. In a meta-analysis including 29 publications, more than half of the studies reported favorable outcomes in oncology patients who received NGS-based testing [8]. Among patients who were matched to targeted treatment, progression-free survival (PFS) was significantly longer in 10 of 14 publications across various tumor types (range of HRs 0.24–0.67, with median of 0.47). Overall survival (OS) was significantly longer in 16 of 26 publications (range of HRs 0.34–0.84, with median of 0.47) [8]. In 16 publications reporting outcomes of patients who received NGS testing, only a median of 25 % (range, 2–66 %) were matched to targeted treatment [8]. The reasons for not receiving matched treatment included therapies were not available, physician chose an alternate treatment, disease has progressed, patient declined treatment, and among others. However, these publications reported outcomes from different types of solid tumors, often mixed in a single cohort. The authors concluded that this makes it difficult to aggregate and compare the impact of CGP across various cancer types [8].
To date, clinical guidelines of the National Comprehensive Cancer Network [52], the European Society for Medical Oncology [53,54], and the American Society of Clinical Oncology recommend genomic testing for some cancers [55]. Similarly, the Asia-Pacific Oncology Drug Development Consortium (APODDC) recommends CGP for patients with NSCLC, epithelial ovarian cancer, or prostate cancer [56].
7. Bridging the standard-of-care to clinical trials
With the ability to profile multiple genes in a single test, CGP helps clinicians evaluate investigational therapies that target rare genetic variants [57]. The "umbrella trial" is designed to study more than one therapy for a particular cancer type defined by both pathological and molecular criteria, which is often guided by CGP nowadays [58]. For example, Lung-MAP is a phase I/II clinical trial conducted in patients with squamous cell lung cancer that aimed to individualize treatment based on the CGP results [59]. This is a multi-institutional, multi-cooperative group trial that aimed to rapidly identify new active drugs and bring them to the patients timely.
Conversely, the "basket trial" is designed to study a therapy targeting particular genes for more than one cancer types [58]. Several large basket trials include the NCI-MATCH, TAPUR, MyPathway, and the ProTarget trials [25,[60], [61], [62]]. Molecular targets and pathways that are frequently targeted in the basket trials include AKT, HER2, HER3, BRAF, Hedgehog, EGFR, BRCA, KIT, PDGFRA, and PDGFRB [63]. Although these genomic alterations can be detected with traditional testing, CGP has greatly facilitated the identification of these variants in a single test, regardless of the prevalence in cancer [64]. Based on CGP, the effectiveness and toxicity of 13 off-labeled targeted therapies in advanced cancers are currently investigated in a phase 2 trial [25].
8. Role of molecular tumor board in the integration of CGP
To facilitate the implementation of CGP into clinical practice, the Molecular Tumor Boards (MTBs) have been established to aid frontline physicians and solve the challenges. MTB usually consists of physicians, pathologists, bioinformaticians, pharmacologists, and many other experts [65]. During the MTB meeting, the physician and/or other members of the care team provide clinical information on the patient, and various scientists provide interpretation of genomic and other molecular data derived from the patient [66]. Subsequently, the participants of the board work together to provide matched treatment options in the form of consensus recommendations [66]. The primary aim of MTB is to provide a multidisciplinary approach to potential therapeutic strategies based on the molecular characteristics of a patient's tumor [67].
In the real-world experience, MTB is a valuable tool for matching CGP data with treatment (in-label use or off-label use) or allocating patients to clinical trials [[68], [69], [70]]. In a meta-analysis study, the percentage of patients receiving MTB-recommended targeted therapies ranged from 11 % to 43 % [71]. Although data quality is limited by a lack of prospective randomized controlled trials, the authors concluded that MTBs appear to improve clinical outcomes for patients with cancer [71]. However, the operation of MTBs usually requires substantial resources and time, indicating that MTBs are not easily scalable [66]. For non-academic institutions or community hospitals, centralization of MTBs might decrease the barrier to promoting treatment decision-making [66]. Most urgently, guidelines for MTB implementation and standardized quality requirements are needed to improve the quality of CGP interpretation for oncology patients.
9. Current scheme of CGP reimbursement policy in Taiwan
Starting from May 2024, Taiwan's National Health Insurance (NHI) has granted the reimbursement of NGS-based CGP for patients with solid cancer and leukemia as part of precision medicine for cancer treatment [72]. The fixed-amount reimbursement encompasses tests for either the BRCA gene, a small panel (≤100 genes), or a large panel (>100 genes), and covers nine solid tumor types. The reimbursement policy also mandates the health-care providers to set up MTBs within or across the institutes. This measure is estimated to benefit over 20,000 cancer patients per year, with a budget of approximately 300 million NTD (about 9 million USD).
Prior to the reimbursement of CGP, the National Biobank Consortium of Taiwan (NBCT) was first inaugurated in 2022 under the auspices of the Ministry of Health and Welfare in Taiwan [73]. The precision oncology branch of the NBCT project was designed to expand the repository of CGP data of Taiwanese oncology patients, fortifying the clinical evidence for precision medicine in this population. As of May 2024, the NBCT project has enrolled 2000 oncology patients and granted tissue-based CGP using the FoundationOne CDx assay [74]. As part of the NBCT project, our experience showed that clinical actionability was detected in 21.1 % (96/456) of the patients enrolled in the study [75]. The clinical outcome of patients who received matched therapy was superior to that of patients who did not receive matched therapy (median survival, 26.1 months and 10.6 months, respectively, hazard ratio of 0.28, 95 % confidence interval of 0.14–0.55, p < 0.001) [75]. The NBCT project also aimed to share the genomic and clinical data of all patients through an integrated platform [76]. Ultimately, the goal of the NBCT project is to facilitate the use of CGP in clinical practice, furnish matched therapies, and eventually improve the therapeutic outcomes and quality of life for cancer patients in Taiwan [76].
10. Challenges of implementing CGP in clinical practice
Despite the advancements of NGS technology and rapid reduction of sequencing cost for the human genome in the past two decades, the cost of CGP remains to be the major barrier for most of the patients to access CGP testing in clinical practice. A national policy is an important enabler for oncology patients to access the CGP. Countries including France, Germany, the United Kingdom, and the United States have introduced national policies designed to increase investments in genomics and increase diagnostic capacity [77]. For patients with metastatic NSCLCs, upfront or sequential (after EGFR and ALK tests) CGP testing was associated with substantial cost savings [78,79]. For patients with advanced gastrointestinal stromal tumor, an uncommon solid cancer harboring somatic KIT or PDGFRA mutations, CGP was associated with an increase of 0.10 quality-adjusted life years (QALYs) at a cost of USD$9513 compared with the empirical imatinib approach [80]. Overall, the economic impact of CGP in other types of malignancies is mostly understudied. To improve the accessibility of CGP for oncology patients, a comprehensive evaluation of the economic impact of CGP is critical to facilitating reimbursement across regions. Other key factors that positively affect NGS implementation in clinical practice include the equipment, infrastructure, routine utilization, expert consultation, and educational programs [81].
Variants of uncertain significance (VUS) are poorly characterized variants in the scientific literature that make their biological and clinical significance unclear [82]. VUSs represent the majority of the variants identified by CGP, account for 60–80 % reported variants in driver genes [82]. These variants are often hard to interpret and leave both patients and physicians perplexed. Recently, machine-learning approaches became an appealing method to predict the pathogenicity of VUS. Tools like AlphaMissense, REVEL, and ESM1b demonstrated high accuracy in predicting the pathogenicity of VUS [[83], [84], [85]]. However, the integration of these variant effect prediction tools into CGP is still at a slow pace.
11. Future direction of CGP in precision oncology
To further profile the cancer cells and provide useful information for oncology patients, a multi-omic approach and data integration from genomics, transcriptomics, epigenomics, proteomics, and microbiomics is a rising trend for precision oncology. More recent developments in tumor profiling enable dissection of tumor molecular architecture and the functional phenotype at cellular and subcellular resolution [86]. In particular, spatial omics are the most promising technologies that combine molecular characterization with spatial resolution, providing unique insights into tumor molecular architecture, intratumor heterogeneity, and tumor microenvironment [86]. Spatial transcriptomic studies were able to dissect intra-tumoral heterogeneity, to understand the tumor-stroma interaction, and to identify biomarkers in lung cancer, colorectal cancer, liver cancer, breast cancer, pancreatic cancer, melanoma, and other cancers [87]. However, clinical translation of high-resolution tumor-stroma profiling and integration of multi-omics data into precision oncology still pose significant challenges. Prospective studies and validations are needed to implement the multi-omic tumor profiling into precision oncology.
12. Conclusions
In the management of oncology patients, the clinical utility of CGP will continue to grow, with broader indication and lower costs over time. Currently, CGP can be applied to DNA and RNA as sequencing material, and performed on tumor tissue and non-tumor tissue. This approach is efficient in identifying actionable targets in cancers and may improve outcomes for oncology patients. CGP also helps to bridge the standard of care with clinical trials, providing opportunities for more treatment options for oncology patients. Consequently, the need for MTB will also increase to support and guide clinicians in interpreting and acting on CGP results. Although it still faces challenges in managing oncology patients, CGP with integrated and multi-omic tumor profiling will probably impact precision oncology in the near future.
References
- 1.Slatko BE, Gardner AF, Ausubel FM. Overview of next-generation sequencing technologies. Curr Protoc Mol Biol. 2018;122 doi: 10.1002/cpmb.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pankiw M, Brezden-Masley C, Charames GS. Comprehensive genomic profiling for oncological advancements by precision medicine. Med Oncol. 2023;41:1. doi: 10.1007/s12032-023-02228-x. [DOI] [PubMed] [Google Scholar]
- 3.Pervez MT, Hasnain MJU, Abbas SH, Moustafa MF, Aslam N, Shah SSM. A comprehensive review of performance of next-generation sequencing platforms. BioMed Res Int. 2022 doi: 10.1155/2022/3457806. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 4.Hung SS, Meissner B, Chavez EA, Ben-Neriah S, Ennishi D, Jones MR, et al. Assessment of capture and amplicon-based approaches for the development of a targeted next-generation sequencing pipeline to personalize lymphoma management. J Mol Diagn. 2018;20:203–214. doi: 10.1016/j.jmoldx.2017.11.010. [DOI] [PubMed] [Google Scholar]
- 5.Singh RR. Target enrichment approaches for next-generation sequencing applications in oncology. Diagnostics. 2022;12 doi: 10.3390/diagnostics12071539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bewicke-Copley F, Arjun Kumar E, Palladino G, Korfi K, Wang J. Applications and analysis of targeted genomic sequencing in cancer studies. Comput Struct Biotechnol J. 2019;17:1348–1359. doi: 10.1016/j.csbj.2019.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Xie N, Shen G, Gao W, Huang Z, Huang C, Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduct Targeted Ther. 2023;8:9. doi: 10.1038/s41392-022-01270-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gibbs SN, Peneva D, Cuyun Carter G, Palomares MR, Thakkar S, Hall DW, et al. Comprehensive review on the clinical impact of next-generation sequencing tests for the management of advanced cancer. JCO Precis Oncol. 2023;7 doi: 10.1200/PO.22.00715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wallenta Law J, Bapat B, Sweetnam C, Mohammed H, McBratney A, Izano MA, et al. Real-world impact of comprehensive genomic profiling on biomarker detection, receipt of therapy, and clinical outcomes in advanced non-small cell lung cancer. JCO Precis Oncol. 2024;8 doi: 10.1200/PO.24.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Espinosa E, Bautista R, Larrosa R, Plata O. Advancements in long-read genome sequencing technologies and algorithms. Genomics. 2024;116 doi: 10.1016/j.ygeno.2024.110842. [DOI] [PubMed] [Google Scholar]
- 11.Sakamoto Y, Miyake S, Oka M, Kanai A, Kawai Y, Nagasawa S, et al. Phasing analysis of lung cancer genomes using a long read sequencer. Nat Commun. 2022;13:3464. doi: 10.1038/s41467-022-31133-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016;17:333–351. doi: 10.1038/nrg.2016.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hu T, Chitnis N, Monos D, Dinh A. Next-generation sequencing technologies: an overview. Hum Immunol. 2021;82:801–811. doi: 10.1016/j.humimm.2021.02.012. [DOI] [PubMed] [Google Scholar]
- 14.Gao Q, Liang WW, Foltz SM, Mutharasu G, Jayasinghe RG, Cao S, et al. Driver fusions and their implications in the development and treatment of human cancers. Cell Rep. 2018;23:227–238. doi: 10.1016/j.celrep.2018.03.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Suda K, Mitsudomi T. Emerging oncogenic fusions other than ALK, ROS1, RET, and NTRK in NSCLC and the role of fusions as resistance mechanisms to targeted therapy. Transl Lung Cancer Res. 2020;9:2618–2628. doi: 10.21037/tlcr-20-186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Heydt C, Wölwer CB, Velazquez Camacho O, Wagener-Ryczek S, Pappesch R, Siemanowski J, et al. Detection of gene fusions using targeted next-generation sequencing: a comparative evaluation. BMC Med Genom. 2021;14:62. doi: 10.1186/s12920-021-00909-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bruno R, Fontanini G. Next generation sequencing for gene fusion analysis in lung cancer: a literature review. Diagnostics. 2020;10 doi: 10.3390/diagnostics10080521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Benayed R, Offin M, Mullaney K, Sukhadia P, Rios K, Desmeules P, et al. High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with No mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden. Clin Cancer Res. 2019;25:4712–4722. doi: 10.1158/1078-0432.CCR-19-0225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Socinski MA, Pennell NA, Davies KD. MET exon 14 skipping mutations in non-small-cell lung cancer: an overview of biology, clinical outcomes, and testing considerations. JCO Precis Oncol. 2021;5 doi: 10.1200/PO.20.00516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, et al. DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J Thorac Oncol. 2019;14:737–741. doi: 10.1016/j.jtho.2018.12.020. [DOI] [PubMed] [Google Scholar]
- 21.Dorney R, Dhungel BP, Rasko JEJ, Hebbard L, Schmitz U. Recent advances in cancer fusion transcript detection. Briefings Bioinf. 2023;24 doi: 10.1093/bib/bbac519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hehir-Kwa JY, Koudijs MJ, Verwiel ETP, Kester LA, van Tuil M, Strengman E, et al. Improved gene fusion detection in childhood cancer diagnostics using RNA sequencing. JCO Precis Oncol. 2022;6 doi: 10.1200/PO.20.00504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Chan HT, Chin YM, Low SK. Circulating tumor DNA-based genomic profiling assays in adult solid tumors for precision oncology: recent advancements and future challenges. Cancers (Basel) 2022;14 doi: 10.3390/cancers14133275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bellosillo B, Montagut C. High-accuracy liquid biopsies. Nat Med. 2019;25:1820–1821. doi: 10.1038/s41591-019-0690-1. [DOI] [PubMed] [Google Scholar]
- 25.Kringelbach T, Højgaard M, Rohrberg K, Spanggaard I, Laursen BE, Ladekarl M, et al. ProTarget: a Danish Nationwide Clinical Trial on Targeted Cancer Treatment based on genomic profiling - a national, phase 2, prospective, multi-drug, non-randomized, open-label basket trial. BMC Cancer. 2023;23:182. doi: 10.1186/s12885-023-10632-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Peters S, Dziadziuszko R, Morabito A, Felip E, Gadgeel SM, Cheema P, et al. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: primary analysis of BFAST cohort C randomized phase 3 trial. Nat Med. 2022;28:1831–1839. doi: 10.1038/s41591-022-01933-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dziadziuszko R, Mok T, Peters S, Han JY, Alatorre-Alexander J, Leighl N, et al. Blood first assay screening trial (BFAST) in treatment-naive advanced or metastatic NSCLC: initial results of the phase 2 ALK-positive cohort. J Thorac Oncol. 2021;16:2040–2050. doi: 10.1016/j.jtho.2021.07.008. [DOI] [PubMed] [Google Scholar]
- 28.Boutonnet A, Pradines A, Mano M, Kreczman-Brun M, Mazières J, Favre G, et al. Size and concentration of cell-free DNA measured directly from blood plasma, without prior DNA extraction. Anal Chem. 2023;95:9263–9270. doi: 10.1021/acs.analchem.3c00998. [DOI] [PubMed] [Google Scholar]
- 29.Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. 2019;25:1928–1937. doi: 10.1038/s41591-019-0652-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Jahangiri L, Hurst T. Assessing the concordance of genomic alterations between circulating-free DNA and tumour tissue in cancer patients. Cancers (Basel) 2019;11 doi: 10.3390/cancers11121938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021;12:11. doi: 10.1038/s41467-020-20162-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Malla M, Loree JM, Kasi PM, Parikh AR. Using circulating tumor DNA in colorectal cancer: current and evolving practices. J Clin Oncol. 2022;40:2846–2857. doi: 10.1200/JCO.21.02615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tu HY, Li YS, Bai XY, Sun YL, Zheng MY, Ke EE, et al. Genetic profiling of cell-free DNA from pleural effusion in advanced lung cancer as a surrogate for tumor tissue and revealed additional clinical actionable targets. Clin Lung Cancer. 2022;23:135–142. doi: 10.1016/j.cllc.2021.09.002. [DOI] [PubMed] [Google Scholar]
- 34.Liu L, Shao D, Deng Q, Tang H, Wang J, Liu J, et al. Next generation sequencing-based molecular profiling of lung adenocarcinoma using pleural effusion specimens. J Thorac Dis. 2018;10:2631–2637. doi: 10.21037/jtd.2018.04.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bae GE, Kim SH, Choi MK, Kim JM, Yeo MK. Targeted sequencing of ascites and peritoneal washing fluid of patients with gastrointestinal cancers and their clinical applications and limitations. Front Oncol. 2021;11 doi: 10.3389/fonc.2021.712754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Han MR, Lee SH, Park JY, Hong H, Ho JY, Hur SY, et al. Clinical implications of circulating tumor DNA from ascites and serial plasma in ovarian cancer. Cancer Res Treat. 2020;52:779–788. doi: 10.4143/crt.2019.700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature. 2009;458:719–724. doi: 10.1038/nature07943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Malone ER, Oliva M, Sabatini PJB, Stockley TL, Siu LL. Molecular profiling for precision cancer therapies. Genome Med. 2020;12:8. doi: 10.1186/s13073-019-0703-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dietel M, Jöhrens K, Laffert MV, Hummel M, Bläker H, Pfitzner BM, et al. A 2015 update on predictive molecular pathology and its role in targeted cancer therapy: a review focussing on clinical relevance. Cancer Gene Ther. 2015;22:417–430. doi: 10.1038/cgt.2015.39. [DOI] [PubMed] [Google Scholar]
- 40.Ossowski S, Neeman E, Borden C, Stram D, Giraldo L, Kotak D, et al. Improving time to molecular testing results in patients with newly diagnosed, metastatic non-small-cell lung cancer. JCO Oncol Pract. 2022;18:e1874–e1884. doi: 10.1200/OP.22.00260. [DOI] [PubMed] [Google Scholar]
- 41.Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017;2017 doi: 10.1200/PO.17.00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lin DI, Quintanilha JCF, Danziger N, Lang L, Levitan D, Hayne C, et al. Pan-tumor validation of a NGS fraction-based MSI analysis as a predictor of response to. Pembrolizumab. npj Precis Oncol. 2024;8:204. doi: 10.1038/s41698-024-00679-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chen KT, Madison R, Moore J, Jin D, Fleischmann Z, Newberg J, et al. A novel HRD signature is predictive of FOLFIRINOX benefit in metastatic pancreatic cancer. Oncologist. 2023;28:691–698. doi: 10.1093/oncolo/oyad178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Antoniotti C, Korn WM, Marmorino F, Rossini D, Lonardi S, Masi G, et al. Tumour mutational burden, microsatellite instability, and actionable alterations in metastatic colorectal cancer: next-generation sequencing results of TRIBE2 study. Eur J Cancer. 2021;155:73–84. doi: 10.1016/j.ejca.2021.06.037. [DOI] [PubMed] [Google Scholar]
- 45.Gouda MA, Nelson BE, Buschhorn L, Wahida A, Subbiah V. Tumor-agnostic precision medicine from the AACR GENIE database: clinical implications. Clin Cancer Res. 2023;29:2753–2760. doi: 10.1158/1078-0432.CCR-23-0090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Le Tourneau C, Delord JP, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334. doi: 10.1016/S1470-2045(15)00188-6. [DOI] [PubMed] [Google Scholar]
- 47.Massard C, Michiels S, Ferté C, Le Deley MC, Lacroix L, Hollebecque A, et al. High-throughput genomics and clinical outcome in hard-to-treat advanced cancers: results of the MOSCATO 01 trial. Cancer Discov. 2017;7:586–595. doi: 10.1158/2159-8290.CD-16-1396. [DOI] [PubMed] [Google Scholar]
- 48.Harttrampf AC, Lacroix L, Deloger M, Deschamps F, Puget S, Auger N, et al. Molecular screening for cancer treatment optimization (MOSCATO-01) in pediatric patients: a single-institutional prospective molecular stratification trial. Clin Cancer Res. 2017;23:6101–6112. doi: 10.1158/1078-0432.CCR-17-0381. [DOI] [PubMed] [Google Scholar]
- 49.Trédan O, Wang Q, Pissaloux D, Cassier P, de la Fouchardière A, Fayette J, et al. Molecular screening program to select molecular-based recommended therapies for metastatic cancer patients: analysis from the ProfiLER trial. Ann Oncol. 2019;30:757–765. doi: 10.1093/annonc/mdz080. [DOI] [PubMed] [Google Scholar]
- 50.Jiao XD, Qin BD, Wang Z, Liu K, Wu Y, Ling Y, et al. Targeted therapy for intractable cancer on the basis of molecular profiles: an open-label, phase II basket trial (Long March Pathway) Front Oncol. 2023;13 doi: 10.3389/fonc.2023.860711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Suehnholz SP, Nissan MH, Zhang H, Kundra R, Nandakumar S, Lu C, et al. Quantifying the expanding landscape of clinical actionability for patients with cancer. Cancer Discov. 2024;14:49–65. doi: 10.1158/2159-8290.CD-23-0467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.National Comprehensive Cancer Network (NCCN) 2024. NCCN clinical practice guidelines in oncology (NCCN Guidelines®)https://www.nccn.org/guidelines/category_1 [Google Scholar]
- 53.Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, 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;31:1491–1505. doi: 10.1016/j.annonc.2020.07.014. [DOI] [PubMed] [Google Scholar]
- 54.Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, 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. 2024;35:588–606. doi: 10.1016/j.annonc.2024.04.005. [DOI] [PubMed] [Google Scholar]
- 55.Chakravarty D, Johnson A, Sklar J, Lindeman NI, Moore K, Ganesan S, et al. Somatic genomic testing in patients with metastatic or advanced cancer: ASCO provisional clinical opinion. J Clin Oncol. 2022;40:1231–1258. doi: 10.1200/JCO.21.02767. [DOI] [PubMed] [Google Scholar]
- 56.Loong HH, Shimizu T, Prawira A, Tan AC, Tran B, Day D, et al. Recommendations for the use of next-generation sequencing in patients with metastatic cancer in the Asia-Pacific region: a report from the APODDC working group. ESMO Open. 2023;8 doi: 10.1016/j.esmoop.2023.101586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Park JJH, Siden E, Zoratti MJ, Dron L, Harari O, Singer J, et al. Systematic review of basket trials, umbrella trials, and platform trials: a landscape analysis of master protocols. Trials. 2019;20:572. doi: 10.1186/s13063-019-3664-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Woodcock J, LaVange LM. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377:62–70. doi: 10.1056/NEJMra1510062. [DOI] [PubMed] [Google Scholar]
- 59.Steuer CE, Papadimitrakopoulou V, Herbst RS, Redman MW, Hirsch FR, Mack PC, et al. Innovative clinical trials: the LUNG-MAP study. Clin Pharmacol Ther. 2015;97:488–491. doi: 10.1002/cpt.88. [DOI] [PubMed] [Google Scholar]
- 60.O’Dwyer PJ, Gray RJ, Flaherty KT, Chen AP, Li S, Wang V, et al. The NCI-MATCH trial: lessons for precision oncology. Nat Med. 2023;29:1349–1357. doi: 10.1038/s41591-023-02379-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Meric-Bernstam F, Hurwitz H, Raghav KPS, McWilliams RR, Fakih M, VanderWalde A, et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): an updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019;20:518–530. doi: 10.1016/S1470-2045(18)30904-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mangat PK, Halabi S, Bruinooge SS, Garrett-Mayer E, Alva A, Janeway KA, et al. Rationale and design of the targeted agent and profiling utilization registry (TAPUR) study. JCO Precis Oncol. 2018;2018 doi: 10.1200/PO.18.00122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hazim A, Prasad V. A pooled analysis of published, basket trials in cancer medicine. Eur J Cancer. 2018;101:244–250. doi: 10.1016/j.ejca.2018.06.035. [DOI] [PubMed] [Google Scholar]
- 64.Tao JJ, Schram AM, Hyman DM. Basket studies: redefining clinical trials in the era of genome-driven oncology. Annu Rev Med. 2018;69:319–331. doi: 10.1146/annurev-med-062016-050343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.El Helali A, Lam TC, Ko EY, Shih DJH, Chan CK, Wong CHL, et al. The impact of the multi-disciplinary molecular tumour board and integrative next generation sequencing on clinical outcomes in advanced solid tumours. Lancet Reg Health West Pac. 2023;36 doi: 10.1016/j.lanwpc.2023.100775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol. 2023;20:843–863. doi: 10.1038/s41571-023-00824-4. [DOI] [PubMed] [Google Scholar]
- 67.Boos L, Wicki A. The molecular tumor board—a key element of precision oncology. memo - Mag Eur Med Onco. 2024;17:190–193. [Google Scholar]
- 68.Vingiani A, Agnelli L, Duca M, Lorenzini D, Damian S, Proto C, et al. Molecular tumor board as a clinical tool for converting molecular data into real-world patient care. JCO Precis Oncol. 2023;7 doi: 10.1200/PO.23.00067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Charo LM, Eskander RN, Sicklick J, Kim KH, Lim HJ, Okamura R, et al. Real-world data from a molecular tumor board: improved outcomes in breast and gynecologic cancers patients with precision medicine. JCO Precis Oncol. 2022;6 doi: 10.1200/PO.20.00508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kato S, Kim KH, Lim HJ, Boichard A, Nikanjam M, Weihe E, 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. doi: 10.1038/s41467-020-18613-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Larson KL, Huang B, Weiss HL, Hull P, Westgate PM, Miller RW, et al. Clinical outcomes of molecular tumor boards: a systematic review. JCO Precis Oncol. 2021;5 doi: 10.1200/PO.20.00495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.National Health Insurance . 2024. Starting from May 1st, the National Health Insurance (NHI) Payment Have Been Covering Next-Generation Sequencing for Solid Cancer and Leukemia as Part of Precision Medicine for Cancer Treatment, Benefitting over 20,000 Patients.https://www.nhi.gov.tw/en/cp-15076-08d24-8-2.html [Google Scholar]
- 73.Feng YA, Chen CY, Chen TT, Kuo PH, Hsu YH, Yang HI, et al. Taiwan Biobank: a rich biomedical research database of the Taiwanese population. Cell Genom. 2022;2 doi: 10.1016/j.xgen.2022.100197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Milbury CA, Creeden J, Yip WK, Smith DL, Pattani V, Maxwell K, et al. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors. PLoS One. 2022;17(3) doi: 10.1371/journal.pone.0264138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hung LJ, Huang CY, Tung KC, Chen JS, Huang WK, Hsu CC, et al. Comprehensive genomic profiling in multiple cancer types: a comparative analysis of the National Biobank Consortium of Taiwan and clinical practice cohorts. J Formos Med Assoc. 2024 doi: 10.1016/j.jfma.2024.09.001. [DOI] [PubMed] [Google Scholar]
- 76.The National Biobank Consortium of Taiwan (NBCT) 2023. Introduction of Cancer Precision Medicine and Biobank Consortium Collaboration Pilot Project.https://nbct.nhri.org.tw/docDetail.aspx?uid=10143&pid=10142&docid=10295&rn=-16307 [Google Scholar]
- 77.Wilsdon T, Horgan D, Akkermans M. Accelerating patient access to next-generation sequencing in oncology: a plan of action. Value & Outcome. 2022;8 [Google Scholar]
- 78.Loong HH, Wong CKH, Chan CPK, Chang A, Zhou ZY, Tang W, et al. Clinical and economic impact of upfront next-generation sequencing for metastatic NSCLC in east Asia. JTO Clin Res Rep. 2022;3 doi: 10.1016/j.jtocrr.2022.100290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pennell NA, Mutebi A, Zhou ZY, Ricculli ML, Tang W, Wang H, et al. Economic impact of next-generation sequencing versus single-gene testing to detect genomic alterations in metastatic non-small-cell lung cancer using a decision analytic model. JCO Precis Oncol. 2019;3:1–9. doi: 10.1200/PO.18.00356. [DOI] [PubMed] [Google Scholar]
- 80.Banerjee S, Kumar A, Lopez N, Zhao B, Tang CM, Yebra M, et al. Cost-effectiveness analysis of genetic testing and tailored first-line therapy for patients with metastatic gastrointestinal stromal tumors. JAMA Netw Open. 2020;3 doi: 10.1001/jamanetworkopen.2020.13565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Horgan D, Van den Bulcke M, Malapelle U, Troncone G, Normanno N, Capoluongo ED, et al. Tackling the implementation gap for the uptake of NGS and advanced molecular diagnostics into healthcare systems. Heliyon. 2024;10 doi: 10.1016/j.heliyon.2023.e23914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mellgard GS, Atabek Z, LaRose M, Kastrinos F, Bates SE. Variants of uncertain significance in precision oncology: nuance or nuisance? Oncologist. 2024;29:641–644. doi: 10.1093/oncolo/oyae135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cheng J, Novati G, Pan J, Bycroft C, Žemgulytė A, Applebaum T, et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science. 2023;381 doi: 10.1126/science.adg7492. [DOI] [PubMed] [Google Scholar]
- 84.Brandes N, Goldman G, Wang CH, Ye CJ, Ntranos V. Genome-wide prediction of disease variant effects with a deep protein language model. Nat Genet. 2023;55:1512–1522. doi: 10.1038/s41588-023-01465-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ioannidis NM, Rothstein JH, Pejaver V, Middha S, McDonnell SK, Baheti S, et al. REVEL: an ensemble method for predicting the pathogenicity of rare missense variants. Am J Hum Genet. 2016;99:877–885. doi: 10.1016/j.ajhg.2016.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Akhoundova D, Rubin MA. Clinical application of advanced multi-omics tumor profiling: shaping precision oncology of the future. Cancer Cell. 2022;40:920–938. doi: 10.1016/j.ccell.2022.08.011. [DOI] [PubMed] [Google Scholar]
- 87.Jin Y, Zuo Y, Li G, Liu W, Pan Y, Fan T, et al. Advances in spatial transcriptomics and its applications in cancer research. Mol Cancer. 2024;23:129. doi: 10.1186/s12943-024-02040-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
