Introduction: The Clinical Imperative for Genomic Interpretation
The practice of oncology is undergoing a fundamental transformation. Historically, cancer was classified and treated based on its anatomical site of origin and histology.1 Today, this paradigm is being supplanted by a molecularly defined approach, where the genetic makeup of a tumor provides a more precise and actionable classification.2 A disease once known simply as "lung cancer" is now understood to be a collection of distinct molecular subtypes, each driven by specific genomic alterations and each potentially susceptible to a different therapeutic strategy.1 This shift presents an unprecedented opportunity for personalized medicine but simultaneously creates a formidable interpretive challenge for the practicing oncologist.3
The widespread clinical adoption of Next-Generation Sequencing (NGS) and Comprehensive Genomic Profiling (CGP) has led to a deluge of complex genomic data.3 Assays like the Pillar OncoReveal CDx can now deliver a detailed genomic report from a tumor sample in under 48 hours, making advanced molecular diagnostics a routine part of patient care.5 However, the technology for generating this data has outpaced the development of widespread clinical infrastructure for its interpretation. This has created a significant "translation gap," where oncologists, despite ordering these powerful tests, often report having educational gaps and lack the specialized training or time to confidently interpret the full spectrum of results, particularly for variants that fall outside of well-established guidelines.7
In this new landscape, two tools have become central to the clinical workflow: the genomic diagnostic report and the curated knowledgebase. The report, generated from the patient's tumor, serves as the primary data input—it is the specific clinical "question" being asked. Curated knowledge bases, such as the Clinical Interpretation of Variants in Cancer (CIViC) database, function as the essential interpretive tool—the "library" where the answer to that question can be found. CIViC's mission is to centralize, debate, and interpret the clinical significance of cancer variants in an open-access, community-driven forum, directly addressing the historical problem of knowledge being siloed in private, "encumbered" databases, which leads to extensive repetition of effort.8 The proliferation of rapid NGS testing has therefore created a new, critical dependency in the clinical workflow: the need for robust, accessible, and continuously updated interpretation tools. These knowledgebases are no longer just academic resources; they are essential clinical infrastructure required to bridge the gap between data generation and clinical action, ensuring the promise of precision medicine can be realized for patients.
Section 1: Deconstructing the Genomic Diagnostic Report: The Oncologist's Starting Point
1.1. The Technology: Targeted NGS Panels in Clinical Practice
The foundation of the modern precision oncology workflow is the targeted NGS panel. These assays use a single test to simultaneously analyze a predefined set of clinically relevant genes, offering a time- and tissue-efficient alternative to sequential single-gene testing.4 A prime example is the Pillar OncoReveal CDx assay, an FDA-approved, in vitro diagnostic (IVD) kit that provides comprehensive genomic results for 22 to 48 key genes implicated in solid tumors.5
From a clinical standpoint, the key features of such panels are their speed and accessibility. With a sample-to-report time of less than 48 hours, these assays provide actionable information within a timeframe that can guide initial treatment decisions for newly diagnosed or progressing patients.5 Furthermore, their design as self-contained kits that can be run on common sequencing platforms like the Illumina MiSeq Dx allows clinical laboratories to bring this testing in-house, granting them greater control over samples and turnaround times rather than relying on external reference labs.6 The Pillar OncoReveal panel holds FDA approval as a companion diagnostic (CDx) for specific, well-established biomarkers—namely, to identify patients with non-small cell lung cancer (NSCLC) who may benefit from EGFR-targeted therapies and patients with colorectal cancer (CRC) who may benefit from KRAS-targeted therapies.5 Beyond these specific CDx claims, it is also approved for General Tumor Profiling (GTP), allowing it to identify a broader range of clinically significant alterations across all solid tumors.5
1.2. Anatomy of the Report: Parsing the Key Sections
The clinical utility of an NGS panel is realized through its final output: the integrated clinical report. This document is carefully structured to present complex genomic data in a hierarchical, clinically intuitive manner. While formats vary, reports from assays like OncoReveal typically contain several key sections that the oncologist must parse.5
- Companion Diagnostic (CDx) Biomarkers: This is the most critical section for immediate decision-making. It lists variants for which there is a direct FDA-approved therapy for the patient's specific cancer type. For example, this section would highlight an EGFR exon 19 deletion in an NSCLC patient or the absence of KRAS codon 12 and 13 mutations (i.e., wild-type) in a CRC patient, directly linking these findings to approved targeted therapies.5
- Mutations with Clinical or Potential Clinical Significance: This section contains the "discovery" findings. It includes other variants detected by the panel that are known to have therapeutic, prognostic, or diagnostic relevance but are not tied to a specific CDx approval for that indication. An example would be finding a BRAF V600E mutation in a colon cancer patient; while this is a highly actionable mutation in melanoma, its implications in colon cancer are different and require further investigation. It is this section that most often necessitates the use of a knowledgebase like CIViC.
- Variants of Uncertain Significance (VUS): These are genetic alterations for which the clinical impact is not yet known. They represent a significant challenge in clinical practice and are generally not used to guide treatment decisions, but may be reclassified over time as more evidence emerges.
- Pertinent "No Calls": This often-overlooked section is crucial for understanding the limitations of the test. It highlights clinically important genes or gene regions that could not be adequately assessed in the patient's sample, perhaps due to low DNA quality or insufficient sequencing coverage.5 This prevents the oncologist from incorrectly assuming a gene is wild-type when, in fact, it was simply not tested.
1.3. The Initial Clinical Triage: From Data to Questions
Upon receiving the genomic report, the oncologist performs an initial triage. The thought process is guided by the report's structure and aims to translate the raw data into a set of actionable clinical questions.
If the report identifies a clear CDx biomarker in the first section—for instance, an EGFR L858R mutation in a patient with NSCLC—the initial treatment path is relatively straightforward and guided by established NCCN guidelines, which recommend specific EGFR inhibitors.10
However, when a variant is listed in the "potential clinical significance" section, the investigative phase begins. The oncologist must formulate a series of precise questions that need to be answered before a treatment plan can be developed:
- Therapeutic Relevance: Does this variant predict sensitivity or resistance to any targeted therapies? This includes therapies approved for other cancer types (off-label use).3
- Prognostic Significance: What does this variant imply about the natural course of the patient's disease, independent of treatment?
- Diagnostic Role: Does this variant help to refine the patient's diagnosis to a more specific molecular subtype?
- Clinical Trial Eligibility: Are there ongoing clinical trials for therapies that specifically target this alteration?
- Germline Potential: Could this somatic finding represent a previously unrecognized germline variant, which would have profound implications for the patient's future cancer risk and for their family members?7
These questions form the precise query that the oncologist will carry forward to a comprehensive knowledge base like CIViC to find evidence-based answers.
Table 1: Anatomy of a Comprehensive Genomic Profiling Report (Pillar OncoReveal CDx Example)
| Section of Report | Description | Clinical Information Provided | Example Finding | Immediate Clinical Question |
|---|---|---|---|---|
| Patient/Sample Information | Details of the patient, tumor type, and sample quality. | Context for the genomic findings. | NSCLC, Adenocarcinoma; 45% Tumor Purity | Is the sample quality adequate for reliable variant detection? |
| Companion Diagnostic (CDx) Results | Variants with direct FDA-approved therapy indications for the specific cancer type. | On-label, standard-of-care treatment options. | EGFR Exon 19 Deletion | Which specific EGFR inhibitor is most appropriate based on NCCN guidelines? |
| Mutations with Clinical Significance | Variants with known clinical relevance that lack a specific CDx claim for this disease. | Potential off-label therapies, prognostic data, or clinical trial options. | PIK3CA H1047R | Does this variant predict response to a PI3K inhibitor in NSCLC? What is the level of evidence? |
| Variants of Uncertain Significance (VUS) | Alterations with unknown clinical impact. | Currently non-actionable information; requires future monitoring. | ATM p.R2443Q | Should this variant be monitored for reclassification in the future? |
| Technical Specifications | Data such as Variant Allele Frequency (VAF), read depth, and assay limitations. | Confidence in the variant call; potential insight into tumor heterogeneity or clonality. | PIK3CA H1047R, VAF: 8% | Is this a subclonal mutation, and how might that affect response to a targeted therapy? |
Section 2: The CIViC Knowledgebase: An Architected Compendium of Clinical Evidence
2.1. Philosophy and Ecosystem: Positioning CIViC
The Clinical Interpretation of Variants in Cancer (CIViC) knowledgebase was created as a direct response to the interpretive challenges of precision oncology. It is founded on the principles of being an open-access, open-source, and community-driven resource.8 This philosophy is crucial, as it aims to break down the information silos of private or proprietary databases, allowing for a transparent, global, and collaborative effort to curate and debate the clinical relevance of cancer variants.14
CIViC occupies a specific and vital niche within the broader ecosystem of genomic databases. While a resource like COSMIC (Catalogue of Somatic Mutations in Cancer) serves as a vast catalog of observed somatic mutations, and ClinVar primarily focuses on the interpretation of germline variants, CIViC's core mission is the detailed curation of literature-derived evidence for the clinical relevance of somatic variants.15 Other valuable resources like OncoKB provide expert panel-driven interpretations and therapeutic actionability levels.15 CIViC's unique contribution is its granular, evidence-based structure, which links every clinical interpretation directly back to a citable publication, promoting transparency and allowing for critical appraisal by the end-user clinician.14
2.2. The Core Data Model: The Building Blocks of Interpretation
To effectively use CIViC, an oncologist must understand its core data model, which organizes information in a hierarchical structure that builds from individual data points to a synthesized clinical consensus.17
- Gene and Variant: These are the most fundamental units, representing the gene of interest (e.g., BRAF) and the specific alteration identified in the report (e.g., V600E).
- Molecular Profile (MP): A pivotal concept in CIViC is that clinical evidence is associated not with a variant alone, but with a Molecular Profile. An MP can be "simple," consisting of a single variant like BRAF V600E. It can also be "complex," representing a combination of two or more variants, such as the co-occurrence of an EGFR mutation and a TP53 mutation. This structure allows the database to capture the clinical significance of more complex genomic contexts.18
- Evidence Item (EID): The EID is the foundational unit of the CIViC knowledgebase. Each EID is a single, structured clinical statement about an MP in a specific disease context, and it is derived from a single, citable source, such as a peer-reviewed publication or a major conference abstract.17 This one-to-one link between an evidence statement and its source is the cornerstone of CIViC's transparency.
- Assertion (AID): An Assertion represents the synthesis of available evidence. It is a higher-level, summary interpretation of the clinical significance of a specific MP in a particular disease, supported by one or more underlying EIDs. Assertions are classified according to established professional guidelines, such as those from the Association for Molecular Pathology (AMP), American Society of Clinical Oncology (ASCO), and College of American Pathologists (CAP), providing a consensus view on the variant's actionability.17
2.3. The Hierarchy of Evidence: The Clinician's Filter for Quality
Perhaps the most critical feature of CIViC for clinical use is its structured hierarchy for weighing evidence. This allows the oncologist to quickly filter vast amounts of information and focus on what is most relevant and robust. This hierarchy is organized along two axes: Evidence Type and Evidence Level.
The Evidence Type defines the nature of the clinical claim being made 14:
- Predictive: The variant's effect on therapeutic response (e.g., sensitivity or resistance to a drug).
- Prognostic: The variant's impact on disease outcome (e.g., overall survival), independent of therapy.
- Diagnostic: The variant's role in classifying a patient with a specific disease or subtype.
- Predisposing: A germline variant's role in conferring susceptibility to developing cancer.
- Functional: Evidence describing how the variant alters the biological function of the protein.
- Oncogenic: Evidence related to the variant's role in tumor pathogenesis.
The Evidence Level describes the strength and type of study that supports the claim, ranging from A (highest) to E (lowest).21 This framework is the primary tool an oncologist uses to differentiate between information that can guide immediate, standard-of-care treatment and information that is merely investigational or hypothesis-generating.
The very structure of the CIViC data model—building from individual Evidence Items to synthesized Assertions, all categorized by type and level—mirrors and reinforces the core principles of evidence-based medicine. The clinical practice of evidence-based medicine requires a physician to find individual studies (the EIDs), critically appraise their quality and relevance (the Evidence Levels and Types), and then synthesize this information to form a clinical judgment or follow a practice guideline (the Assertion). By using CIViC, an oncologist is not simply looking up a fact; they are guided through a digitized, transparent process of this exact reasoning. The platform’s design implicitly teaches the user how to think critically about genomic evidence, transforming a simple database query into a practical exercise in evidence-based medicine.
Table 2: The CIViC Evidence Hierarchy: A Clinician's Guide to Weighing Genomic Data
| Level | Level Name | Definition | Typical Source | Clinical Implication & Actionability |
|---|---|---|---|---|
| A | Validated Association | Proven/consensus association in human medicine. | FDA approvals, NCCN/ASCO guidelines, Phase III clinical trials. | Supports on-label, guideline-adherent therapy. High confidence for clinical decision-making. |
| B | Clinical Evidence | Association supported by primary patient data from multiple patients. | Phase I/II clinical trials, large cohort studies, retrospective analyses. | May support off-label consideration, strong rationale for clinical trial referral. |
| C | Case Study | Observations from individual case reports or a very small series of patients (<5). | Published case reports in clinical journals. | Hypothesis-generating only. Not sufficient for guiding therapy outside of a clinical trial. |
| D | Preclinical Evidence | Association supported by in vivo or in vitro models. | Cell line experiments, animal (e.g., mouse) models. | Establishes biological plausibility. Does not directly support clinical action. |
| E | Inferential Association | Indirect evidence where the association is at least one step removed from direct observation. | In silico predictions, correlative studies without direct functional data. | Weakest level of evidence. Not for clinical use. |
Table 3: Comparative Overview of Key Oncology Knowledge bases
| Feature | CIViC | OncoKB | MyCancerGenome / CGI |
|---|---|---|---|
| Curation Model | Open, community-driven, expert-moderated crowdsourcing.[8, 14] | Expert panel curation from a single institution (Memorial Sloan Kettering).[15, 22] | Expert panel curation from a multi-institutional consortium.[23] |
| Primary Focus | Granular, literature-derived evidence items linking variants to clinical outcomes.17 | Therapeutic actionability levels, oncogenicity, and FDA/NCCN guideline information.15 | Comprehensive information on biomarkers, therapies, and clinical trials.[23] |
| Evidence Tiering System | Evidence Levels A-E (study type) and Evidence Types (Predictive, Prognostic, etc.).21 | OncoKB Levels of Evidence (e.g., Level 1, 2, 3A, 3B, 4, R1, R2) for therapies.[23] | Tiered evidence based on clinical guidelines and study strength.[24] |
| Access Model | Fully open access and open source (CC0 license).[8, 25] | Free for academic/research use; requires license for commercial use.[22] | Free for academic/research use; registration required.[23] |
| Linkage to Guidelines | Assertions can link to NCCN guidelines and FDA approvals.17 | Directly incorporates and displays NCCN guideline recommendations and FDA labels.[26] | Provides links and summaries related to clinical guidelines. |
Section 3: The Clinical Workflow: A Step-by-Step Guide from Query to Interpretation
To illustrate the practical application of this workflow, consider a hypothetical case: a 62-year-old patient with newly diagnosed, metastatic NSCLC of the adenocarcinoma subtype. The Pillar OncoReveal report returns, showing no common EGFR, ALK, or ROS1 alterations, but it does identify a MET Exon 14 Skipping Mutation. The oncologist now turns to CIViC to interpret this finding.
3.1. Step 1: Targeted Information Retrieval in CIViC
The process begins with a targeted search. The oncologist uses the main search bar on the CIViC website to query the gene of interest: "MET".17 This leads to the main gene page for MET, which provides a curated summary of the gene's overall clinical relevance. From this page, the oncologist can browse the list of associated variants and Molecular Profiles to find the specific alteration from the report: "Exon 14 Skipping". Clicking on this link takes them to the dedicated Molecular Profile page, which is the central hub for all curated evidence related to this specific variant.
3.2. Step 2: Critical Appraisal of Evidence Items (EIDs)
The Molecular Profile page for MET Exon 14 Skipping presents a list of all associated EIDs. To make sense of this information, the oncologist employs a systematic process of filtering and sorting.
First, they filter by Evidence Type. Since the primary clinical question is therapeutic, the oncologist selects "Predictive" to narrow the results to only those EIDs that describe a response or resistance to therapy.
Next, they sort the filtered results by Evidence Level, from A down to E. This immediately brings the most robust and clinically relevant information to the top. The oncologist will find multiple Level A and Level B EIDs. These EIDs will describe pivotal clinical trials, such as the GEOMETRY mono-1 study for capmatinib and the VISION study for tepotinib, which demonstrated significant and durable responses to these MET inhibitors in patients with NSCLC harboring MET exon 14 skipping mutations.21 The EID summary provides a concise description of the study, including the number of patients, the objective response rate (ORR), and statistical significance.17 Critically, each EID also provides a direct link to the source publication in PubMed and, where applicable, the clinical trial identifier (NCT number), allowing for a seamless transition to the primary data if needed.28 Further down the list, the oncologist might see Level C (case study) or Level D (preclinical) evidence, which they note but recognize as not being actionable for the immediate treatment decision.
3.3. Step 3: Synthesizing Consensus with Assertions (AIDs)
After reviewing the key individual pieces of evidence, the oncologist navigates to the "Assertions" tab for the MET Exon 14 Skipping Molecular Profile. This view provides the synthesized, consensus interpretation.
Here, they will find a Predictive Assertion for this variant in the context of NSCLC. The Assertion Summary will provide a clear, top-line statement, such as: " MET Exon 14 Skipping predicts sensitivity to MET inhibitors in Non-Small Cell Lung Cancer." The Assertion Description offers a narrative summary of the supporting evidence, referencing the key clinical trials. Most importantly, the Assertion will display the formal classification according to professional guidelines: AMP/ASCO/CAP Tier I, Level A.17 This classification provides definitive confirmation that the variant has strong, validated clinical significance and is associated with FDA-approved therapies recommended by professional guidelines. The Assertion may also feature direct links to the relevant FDA companion diagnostic approval and the NCCN Guidelines, creating a direct bridge from the curated evidence to the established standards of care.17
3.4. Step 4: Exploring the Therapeutic Frontier (On- and Off-Label)
The findings from the previous steps solidify the treatment path for this patient. The Tier I, Level A Assertion confirms that treating this patient's NSCLC with a MET inhibitor like capmatinib or tepotinib is an on-label, standard-of-care therapeutic option.
This workflow also demonstrates how CIViC supports decision-making in more ambiguous scenarios. If the variant in question had been a rarer MET mutation with only Level B or C evidence and no Tier I Assertion, the process would be different. In that case, the oncologist would use the EIDs to identify therapies that have shown promise but are not yet approved for this specific context. They could then use the linked NCT numbers to explore clinicaltrials.gov and determine if the patient might be eligible for enrollment in a study investigating a MET inhibitor for their specific rare mutation.28 This highlights CIViC's utility not only in confirming standard-of-care but also in navigating the frontier of investigational medicine and clinical trial opportunities.
This clinical workflow is a dynamic process of adjusting informational resolution. The oncologist can "zoom out" to get a quick, high-level verdict from the Assertion page—the Tier I/A classification answers the immediate question of actionability. However, to practice medicine responsibly, they must also be able to "zoom in" to the underlying EIDs to understand the specific trials, patient populations, and effect sizes that form the basis of that high-level recommendation. If the case is particularly complex or the evidence is conflicting, they can "zoom in" even further by clicking through to the primary PubMed source. Finally, armed with this multi-layered understanding, they "zoom out" again to formulate the treatment plan. This ability to modulate the depth of inquiry based on the clinical question's complexity is what makes a well-structured knowledgebase an indispensable tool.
Section 4: Clinical Integration and Final Treatment Formulation
4.1. Harmonization with Professional Guidelines
Information gleaned from a knowledgebase like CIViC serves as powerful decision support, but it must be validated against the gold standards of clinical practice. The final therapeutic decision is not made in a vacuum; it is made in the context of established, expert-vetted professional guidelines.
The oncologist's next step is to consult the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer.12 Within the guideline document, they will navigate to the algorithms for molecular testing and treatment of metastatic disease. The NCCN Guidelines will explicitly recommend testing for MET exon 14 skipping mutations and will list capmatinib and tepotinib as "Category 1" or "Preferred" first-line treatment options for patients whose tumors harbor this alteration.31 This step provides external validation of the findings from CIViC and confirms that the proposed treatment aligns with the national standard of care. The AMP/ASCO/CAP guidelines provide the framework for how variants are classified (the Tier I-IV system), and seeing this same system used within CIViC confirms that the knowledgebase and the clinical lab are speaking the same standardized language of evidence.20
4.2. The Role of the Molecular Tumor Board (MTB)
For a clear-cut, guideline-concordant case like MET Exon 14 Skipping in NSCLC, a formal Molecular Tumor Board discussion might be brief or even unnecessary. However, the MTB is a critical component of the precision oncology ecosystem, particularly for more complex cases.32 If the genomic report had revealed multiple potentially actionable variants, or if the evidence from CIViC had been ambiguous (e.g., conflicting Level B EIDs with no Tier I Assertion), the case would be presented to the MTB.
In an MTB meeting, the treating oncologist would present the patient's clinical history, the genomic report, and the evidence they gathered from CIViC and other knowledge bases. A multidisciplinary team—including medical oncologists, pathologists, genetic counselors, bioinformaticians, and pharmacists—would then collectively review and debate the evidence. This collaborative process leverages diverse expertise to arrive at a consensus recommendation, which has been shown to improve outcomes for patients receiving genomically matched therapies.32
4.3. The Final Synthesis: Crafting the Patient-Specific Plan
The final step in the workflow is the synthesis of all available information into a tailored treatment plan for the individual patient. This involves integrating four key domains of information:
- Genomic Data: The patient’s tumor has a MET Exon 14 Skipping mutation.
- Curated Evidence (CIViC): A Tier I, Level A Assertion, supported by strong Level A/B EIDs, confirms the variant's predictive value for MET inhibitors.
- Clinical Guidelines (NCCN): The standard of care for this molecular subtype of NSCLC is treatment with capmatinib or tepotinib.
- Patient-Specific Factors: The oncologist must now consider the individual patient. This includes their age, performance status, comorbidities (e.g., renal or hepatic function), potential drug-drug interactions, and the specific toxicity profiles of the available drugs (e.g., peripheral edema is a known side effect of MET inhibitors).
After weighing all these factors, the oncologist meets with the patient. They recommend initiating treatment with capmatinib, explaining the rationale in clear terms: the treatment is chosen not just for "lung cancer," but specifically because the tumor has a particular genetic "switch" that this drug is designed to turn off. They can communicate the high level of confidence in this choice, backed by major clinical trials and national guidelines. This comprehensive, evidence-based process ensures the final recommendation is not just genomically informed, but also clinically appropriate and patient-centered.
This entire workflow, from receipt of the report to the final treatment recommendation, can be viewed as a formal process of risk mitigation. Each step is designed to reduce uncertainty and the potential for error in a high-stakes clinical decision. The internal quality controls of the NGS assay provide the first layer of safety.5 Using a knowledgebase like CIViC mitigates the risk of acting on a single, potentially misinterpreted study by aggregating and tiering all available evidence.17 Cross-referencing with NCCN guidelines mitigates the risk of deviating from the established, expert-vetted standard of care.12 Discussion at an MTB mitigates the risk of individual cognitive bias by leveraging collective expertise.32 Finally, considering patient-specific factors mitigates the risk of applying a guideline-concordant therapy to a patient who cannot tolerate it. Precision oncology is often framed as finding the "right drug," but this workflow demonstrates that the practice is equally about building a robust, multi-layered "safety case" for each treatment decision.
Conclusion: The Indispensable Role of Curated Knowledge in an Era of Data-Driven Medicine
The journey from a variant identified on a genomic report to a final verdict in a patient's treatment plan is a sophisticated, multi-step process of inquiry, appraisal, and synthesis. It demands that the modern oncologist act not only as a clinician but also as an applied informatician, capable of navigating complex data landscapes to find actionable evidence. This report has detailed a systematic workflow that begins with the deconstruction of a genomic report, proceeds through a structured query and critical appraisal of evidence within the CIViC knowledgebase, and concludes with the integration of these findings with professional guidelines and patient-specific factors to formulate a sound therapeutic strategy.
Open-access, transparent, and expertly curated knowledgebases like CIViC have become indispensable clinical infrastructure in the era of data-driven medicine. They empower individual clinicians with the synthesized knowledge of the global research community, standardize the language and criteria for variant interpretation across institutions, and ultimately, facilitate the delivery of truly personalized and evidence-based cancer care.8 As the volume and complexity of genomic data continue to grow, the reliance on such resources will only intensify. Future challenges will include managing the ever-expanding list of variants of uncertain significance and integrating advanced computational tools, such as artificial intelligence and machine learning, to augment and accelerate this expert-driven clinical workflow.34 However, the core principles of transparent evidence curation and critical clinical appraisal, as embodied by the workflow described, will remain the bedrock of responsible precision oncology.
Works cited
- Spotlight on comprehensive genomic profiling - Roche, accessed October 30, 2025, https://www.roche.com/stories/comprehensive-genomic-profiling-in-personalised-healthcare
- Genomic Profiling, accessed October 30, 2025, https://nlmsf.org/genomic-profiling/
- Personalized cancer therapy—leveraging a knowledge base for clinical decision-making, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5880252/
- Comprehensive Genomic Profiling (CGP) | Cancer genomic profiling benefits - Illumina, accessed October 30, 2025, https://www.illumina.com/areas-of-interest/cancer/ngs-in-oncology/cgp.html
- oncoReveal CDx - Get More Patients on Therapy. - Pillar Biosciences, accessed October 30, 2025, https://www.pillarbiosci.com/wp-content/uploads/2024/04/MK-0051-OncoReveal-CDx-FDA-Product-Sheet-4.22.2024-v1.1.pdf
- Pillar Biosciences' FDA-Approved Solid Tumor NGS Kit oncoReveal™ CDx Launched at Molecular Pathology Laboratory Network, Inc., accessed October 30, 2025, https://www.mplnet.com/pillar-biosciences-fda-approved-solid-tumor-ngs-kit-oncoreveal-cdx/
- Oncologists' Perceptions of Tumor Genomic Profiling and the Communication of Test Results and Risks - Karger Publishers, accessed October 30, 2025, https://karger.com/phg/article/24/5-6/304/826335/Oncologists-Perceptions-of-Tumor-Genomic-Profiling
- CIViC - Clinical Interpretation of Variants in Cancer, accessed October 30, 2025, https://civicdb.org/pages/about
- Pillar oncoReveal Solid Tumor v2 Panel | Oncology NGS panel - Illumina, accessed October 30, 2025, https://www.illumina.com/products/by-type/sequencing-kits/library-prep-kits/pillar-oncoreveal-solid-tumor.html
- oncoReveal™ Dx Lung and Colon Cancer Assay - Pillar Biosciences, accessed October 30, 2025, https://www.pillarbiosci.com/wp-content/uploads/2021/07/MK-0027-2-IVD-Product-Sheet-EU.pdf
- Clinical application of a cancer genomic profiling assay to guide precision medicine decisions - PMC - PubMed Central, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5580078/
- Biomarkers Compendium - NCCN, accessed October 30, 2025, https://www.nccn.org/compendia-templates/compendia/biomarkers-compendium
- Establishment of a comprehensive set of fact sheets for cancer predisposition genes for medical oncologists practicing cancer genome profiling - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12014809/
- (PDF) CIViCdb 2022: evolution of an open-access cancer variant interpretation knowledgebase - ResearchGate, accessed October 30, 2025, https://www.researchgate.net/publication/365370639_CIViCdb_2022_evolution_of_an_open-access_cancer_variant_interpretation_knowledgebase
- Chapter 6 Introduction to Cancer Variant Knowledgebases, accessed October 30, 2025, https://course.civicdb.org/introduction-to-cancer-variant-knowledgebases.html
- Comparison of Open-access Databases for Clinical Variant Interpretation in Cancer: A Case Study of MDS/AML, accessed October 30, 2025, https://cgp.iiarjournals.org/content/18/2/157/tab-figures-data
- Introduction to CIViC — CIViC documentation, accessed October 30, 2025, https://civic.readthedocs.io/en/latest/
- Prioritizing Curation Effort — CIViC documentation - Read the Docs, accessed October 30, 2025, https://civic.readthedocs.io/en/latest/curating/ideas.html
- Clinical Interpretation of Variants in Cancer: CIViC, accessed October 30, 2025, https://civicdb.org/
- 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 - PubMed, accessed October 30, 2025, https://pubmed.ncbi.nlm.nih.gov/27993330/
- Evidence Items — CIViC documentation, accessed October 30, 2025, https://civic.readthedocs.io/en/latest/model/evidence.html
- Clinical Trials Home | CIViC, accessed October 30, 2025, https://civicdb.org/clinical-trials
- Clinical Trial — CIViC documentation - Read the Docs, accessed October 30, 2025, https://civic.readthedocs.io/en/latest/model/evidence/clinical_trial.html
- A Simplified Approach to Somatic Analysis in NGS - Euformatics, accessed October 30, 2025, https://www.euformatics.com/blog-post/a-simplified-approach-to-somatic-analysis-in-ngs
- Clinical Trials, accessed October 30, 2025, https://clinicaltrials.gov/
- Detection, Prevention, and Risk Reduction - NCCN, accessed October 30, 2025,
- Usefulness of Comprehensive Genomic Profiling in Clinical Decision-Making in Oncology: A Systematic Review - Allen Press, accessed October 30, 2025, https://meridian.allenpress.com/innovationsjournals-JIPO/article/8/1/55/505025/Usefulness-of-Comprehensive-Genomic-Profiling-in
- CIViCutils: Matching and downstream processing of clinical annotations from CIViC., accessed October 30, 2025, https://f1000research.com/articles/12-1304
- Press Release Service: Genomic Cancer Panel and Profiling Market Research Report 2025-2029 with Executive & Consultant Guides - ResearchAndMarkets.com, accessed October 30, 2025, https://crisprmedicinenews.com/press-release-service/card/genomic-cancer-panel-and-profiling-market-research-report-2025-2029-with-executive-amp-consultant/
- Comparison to OncoKB and CIViC Upset plots comparing PubMed ids,... - ResearchGate, accessed October 30, 2025, https://www.researchgate.net/figure/Comparison-to-OncoKB-and-CIViC-Upset-plots-comparing-PubMed-ids-therapies-and-genes_fig4_354973543