Contacts
Contact Us
Close

Contacts

7505 Fannin St.
Suite 610
Houston, TX 77054

+1 (713) 489-9827

partnerships@biostate.ai

RNA Fusion in Cancer Detection and Analysis

RNA Fusion in Cancer Detection and Analysis

In precision medicine, identifying cancer-driving mutations is transforming the way we diagnose and treat cancers. RNA fusions, a type of genetic alteration where two genes unexpectedly join, have become a crucial part of cancer research and clinical oncology.

Advances in next-generation sequencing have improved the ability to detect these fusions, providing insights that help guide targeted therapies and personalized patient care. A 2024 study revealed that RNA fusion panels clarified or refined diagnoses in over 24.3% of solid tumor cases, with success rates surpassing 93% in clinical settings.

This article will explore why RNA fusions are becoming central to cancer diagnostics, the latest detection technologies, and how their integration is driving innovation in the pharmaceutical and biotech industries.

Key Takeaways

  • RNA fusions play a crucial role in cancer detection by serving as biomarkers that identify driver mutations, guide personalized treatments, and predict patient prognosis. 
  • These genetic alterations happen when two genes fuse, creating a chimeric RNA molecule that produces a fusion protein, which drives cancer progression. 
  • RNA sequencing (RNA-seq) advancements have improved fusion detection, offering better sensitivity and specificity compared to traditional methods like RT-PCR and FISH.
  • Although challenges like RNA degradation and expression-dependent detection remain, innovations such as liquid biopsies, long-read sequencing, and AI-powered analysis are enhancing RNA fusion detection, solidifying it as a key component in precision oncology.

What are RNA Fusions and Why Do They Matter in Cancer?

RNA fusions occur when two separate genes join together, creating a new RNA molecule that codes for a fusion protein. Chromosomal changes, such as translocations, inversions, or deletions, cause these fusions. When DNA breaks and incorrectly rejoins, it forms a chimeric RNA with sequences from both genes.

How RNA Fusions Form

Chromosomal rearrangements often cause RNA fusions. These changes lead to DNA breaks at specific points, causing parts of two genes to join incorrectly. The result is a fused RNA molecule.

Why RNA Fusions Matter in Cancer

Why RNA Fusions Matter in Cancer

RNA fusions matter for several reasons:

  • Driver Mutations: Many RNA fusions create proteins that drive cancer. These fusion proteins gain new functions or lose regulation, promoting uncontrolled cell growth. For example, the BCR-ABL1 fusion in leukemia creates a protein that drives cell proliferation.
  • Diagnostic Markers: Specific RNA fusions act as molecular signatures, helping doctors diagnose and classify cancers. The EWSR1-FLI1 fusion marks Ewing sarcoma.
  • Therapeutic Targets: Fusion proteins often remain unique to cancer cells, making them prime drug targets. The success of imatinib for BCR-ABL1-positive cancers demonstrates how targeting fusion proteins can lead to effective therapies.
  • Prognostic Indicators: Some RNA fusions predict how well patients will respond to treatment. Others signal more aggressive cancer behavior.

Clinical Examples

Fusion genes have transformed cancer treatment. The EML4-ALK fusion in lung cancer responds well to ALK inhibitors like crizotinib. NTRK fusions found across various cancers respond to TRK inhibitors like larotrectinib. These targeted therapies often work better and cause fewer side effects than traditional chemotherapy.

RNA fusions play a vital role in cancer biology and precision medicine, offering valuable insights and treatment options. As sequencing technology improves, doctors will continue to expand treatment choices by targeting these fusions.

How RNA Fusions Drive Cancer Progression

RNA fusions, or chimeric RNAs, are key drivers of cancer. These molecules play a major role in turning normal cells into cancerous ones. They form through various biological processes and contribute to tumor growth, drug resistance, and metastasis.

How RNA Fusions Form

RNA fusions can form in two main ways: chromosomal rearrangements and splicing mechanisms. Chromosomal rearrangements, such as translocations and deletions, join two genes at the DNA level, creating chimeric RNA. This is common in sarcomas and blood cancers.

RNA fusions can also form through splicing without genomic changes. In trans-splicing, exons from different chromosomes join. In cis-splicing, adjacent genes undergo splicing within 30,000 base pairs. Surprisingly, 18% of fusion RNAs show no genomic rearrangement, highlighting the role of splicing in fusion formation.

Role of RNA Fusion in Cancer Progression

RNA fusions drive cancer by altering key cell functions and promoting cancer development.

Kinase Activation and Signaling Disruption

Many RNA fusions involve kinase genes. These fusions create active kinases that promote cell growth and survival. Examples include BCR-ABL1 in leukemia and ALK fusions in lung cancer. These fusions activate important pathways like RTK-RAS, PI3K-AKT, and MAPK, which drive cancer growth.

Transcriptional Changes

RNA fusions involving transcription factors also contribute to cancer. These fusion proteins can disrupt normal gene regulation, leading to abnormal cell behavior. The PAX3-FOXO1 fusion in rhabdomyosarcoma creates a transcription factor that drives tumor growth.

Promoter hijacking is another key process. Strong promoters can drive overexpression of oncogenes, like in TMPRSS2-ERG fusion in prostate cancer.

Non-coding RNA Functions

Some RNA fusions do not produce proteins. Instead, they act as non-coding RNAs, which regulate gene expression through mechanisms like epigenetic changes and microRNA interactions. These non-coding fusion RNAs play an important role in cancer progression.

Impact on Cancer Development

RNA fusions contribute to multiple cancer hallmarks, helping tumors grow and spread.

Cellular Plasticity and Differentiation

Chimeric RNAs help maintain cancer stem cell properties and promote dedifferentiation, allowing cancer cells to return to a more primitive state. This disruption can prevent normal differentiation and aid in cancer progression.

Drug Resistance

RNA fusions contribute to drug resistance. Fusion kinases help cancer cells survive DNA-damaging treatments and promote survival signaling. Additionally, fusion RNAs generate diversity, helping cancer cells adapt to therapy.

Tumor Microenvironment Modulation

RNA fusions also influence the tumor microenvironment. Fusion proteins can alter exosome content, promoting communication between tumor cells and their environment, which can enhance metastasis.

Therapeutic Implications

Recognizing RNA fusions has led to new cancer treatments, especially targeted therapies.

Targeted Therapies

Kinase fusions are ideal targets for small-molecule inhibitors. Imatinib for BCR-ABL1, crizotinib for ALK fusions, and larotrectinib for NTRK fusions have all shown great success in treating cancers with these fusions.

Diagnostic and Prognostic Use

RNA fusion detection is crucial for diagnosing and predicting cancer outcomes. Panels that detect multiple fusions are now used in clinical practice, identifying actionable fusion events in 10-20% of patients.

New Therapeutic Approaches

New Therapeutic Approaches

In addition to kinase inhibitors, new therapies include:

  • Genome editing: Targeting specific fusion breakpoints using advanced CRISPR systems.
  • Immunotherapies: Targeting fusion-specific neoantigens.
  • Combination therapies: Addressing multiple pathways activated by fusion proteins.

RNA fusions are critical to cancer progression. They activate kinases, disrupt gene regulation, and promote drug resistance. As detection methods improve, RNA fusions will continue to play a vital role in precision cancer treatment, offering new opportunities for diagnosis, prognosis, and targeted therapies.

By integrating fusion detection into clinical practice, we can significantly improve cancer treatment and patient outcomes.

How RNA Fusion Can Detect Cancer?

RNA fusions serve as powerful biomarkers for cancer detection, providing both diagnostic and prognostic insights. These chimeric RNA molecules act as specific markers of malignancy and can be detected through advanced molecular techniques. This has revolutionized early cancer detection and monitoring.

RNA Fusions as Cancer Biomarkers

RNA fusions are ideal biomarkers for cancer because they are typically absent in normal tissues but highly expressed in tumors. Unlike DNA alterations, which may not be actively transcribed, RNA fusions reflect oncogenic gene expression, making them crucial for cancer detection. They help pinpoint tumor types and guide targeted therapies.

Recent studies show that 42.7% of solid tumors harbor detectable RNA fusion events. These events lead to diagnostic clarification or refinement in 31.9% of cases, underscoring the importance of RNA fusion detection in cancer diagnosis.

Detection Methods and Technologies

Next-Generation Sequencing (NGS)

Targeted RNA sequencing has become the gold standard for detecting fusions. It offers greater sensitivity and specificity compared to traditional methods. RNA-seq can detect multiple fusion genes, provide quantitative expression data, and identify new fusion partners.

In pediatric cancers, RNA sequencing increased diagnostic yield by 38-39%, detecting fusion events that traditional methods missed. RNA-seq achieved 100% sensitivity and specificity in validation studies.

Comparison with Traditional Methods

RNA fusion detection outperforms traditional methods in key areas:

MethodSensitivitySpecificityAdvantagesLimitations
RNA-seq NGS95-100%99-100%Comprehensive, quantitative detectionHigher cost, requires expertise
RT-PCR62-100%94-100%Fast, cost-effective, minimal tissueLimited to known fusions
FISHVariable85-98%Direct genomic detectionLower sensitivity, labor-intensive
IHCVariableVariableProtein detectionVariable interpretation

RT-PCR shows 100% sensitivity with optimal conditions, but drops at stricter thresholds. RNA-seq detects fusions with as few as 50 transcript copies and works well in challenging FFPE samples.

Long-Read Sequencing Advances

Long-read sequencing improves fusion detection by offering higher resolution for complex fusion events. These technologies help detect fusion isoforms missed by short-read methods, providing complete fusion transcript structures.

Studies show long-read RNA sequencing improves sensitivity, especially for complex rearrangements with multiple breakpoints or unusual splicing patterns.

Liquid Biopsy Applications

Circulating Cell-Free RNA

Liquid biopsies using circulating cell-free RNA (cfRNA) offer a non-invasive way to detect cancer. Studies show 95% accuracy in detecting early-stage colorectal cancer with cfRNA-based assays.

In lung cancer, cfRNA fusion detection showed 77.8% sensitivity, outperforming ctDNA detection, which had 33.3% sensitivity in treatment-naïve patients.

Exosomal RNA Detection

Exosomal RNA detection has become a promising liquid biopsy technique. Exosomes protect RNA, offering stable biomarkers. Microfluidic technologies now allow ultrafast exosomal RNA detection in 10 minutes, requiring just 30 μL of serum.

Circulating Tumor Cells

Circulating tumor cells (CTCs) containing fusion transcripts provide insights into tumor heterogeneity and metastatic potential. CTC-based fusion detection allows real-time monitoring of disease progression.

Multi-Analyte Liquid Biopsy

The future of fusion-based cancer detection lies in comprehensive liquid biopsy platforms that analyze multiple biomarkers, including cfRNA, CTCs, and exosomal RNA, improving detection sensitivity and providing full tumor profiling.

Point-of-Care Testing

Point-of-care fusion detection systems could revolutionize cancer screening, making advanced diagnostics available in diverse clinical settings. Microfluidic technologies can complete fusion RNA analysis in minutes instead of hours.

Personalized Monitoring

Patient-specific fusion monitoring systems will guide personalized cancer surveillance and treatment. These systems will detect early recurrence and guide treatment decisions with unprecedented precision.

Advanced Detection Technologies

Digital PCR

Digital PCR (dPCR) enhances fusion detection sensitivity and provides absolute quantification without needing standard curves. It excels in minimal residual disease monitoring and detecting low-abundance fusion transcripts.

Artificial Intelligence Integration

AI and machine learning improve fusion detection by analyzing complex sequencing data. These tools increase accuracy, reduce false positives, and automate result interpretation.

RNA fusion detection is transforming cancer diagnosis and monitoring. With high sensitivity and specificity, RNA fusion analysis enables earlier detection, precise diagnosis, and targeted therapies. As detection methods improve and costs decrease, RNA fusion analysis will become central to precision cancer medicine, offering new opportunities for early detection, accurate diagnosis, and personalized treatments. The integration of AI, liquid biopsies, and point-of-care technologies will further enhance its clinical utility, improving cancer outcomes worldwide.

Sample Types and Technical Considerations for RNA Fusion in Cancer Detection 

Sample Types for RNA Fusion in Cancer Detection

Fresh and Frozen Tissues

  • Fresh and frozen tissues are the gold standard for RNA fusion detection due to their high RNA quality. Fresh tissue provides the best RNA, ideal for fusion detection, but it’s often not available in clinical practice.
  • Frozen tissues, stored at -80°C, maintain RNA quality well. They show high correlation with fresh tissue in gene expression and fusion detection, making them a reliable alternative.

Formalin-Fixed Paraffin-Embedded (FFPE) Tissues

FFPE tissues are the most common sample type in pathology archives, useful for retrospective studies and diagnostics. However, formalin fixation can degrade RNA, making fusion detection challenging.

FFPE samples often show:

  • Lower RNA quality with degraded fragments.
  • More unmapped reads due to RNA fragmentation.
  • Reduced representation of protein-coding genes.

Despite these challenges, FFPE tissues can still yield useful fusion detection results if proper protocols are followed.

Liquid Biopsy Samples

Circulating cell-free RNA (cfRNA) and exosomal RNA from blood offer non-invasive options for fusion detection. These samples provide:

  • Non-invasive collection, ideal for monitoring.
  • Real-time disease assessment without tissue biopsy.
  • Potential for early detection before symptoms appear.

However, these samples contain low RNA concentrations and need specialized protocols for fusion detection.

RNA Quality Assessment and Technical Considerations

Two key metrics assess RNA quality: RNA Integrity Number (RIN) and DV200 index.

RNA Integrity Number (RIN)

RIN ranges from 1 (completely degraded) to 10 (intact RNA) and is commonly used for RNA quality assessment. However, it is less reliable for FFPE samples and doesn’t always correlate with next-gen sequencing (NGS) performance.

DV200 Index

The DV200 index measures the percentage of RNA fragments longer than 200 nucleotides. It’s better for NGS and correlates well with library yield and FFPE sample quality. A DV200 ≥30% is considered the threshold for acceptable RNA quality for fusion detection.

Impact of RNA Degradation on Fusion Detection

RNA degradation affects fusion detection, especially with poly-A pulldown methods. Degraded RNA shows decreased coverage from the 3′ end of genes, making fusion detection less sensitive for breakpoints farther from this end.

For instance:

  • 95% detection at 1 kb from the 3′ end.
  • 45% at 3 kb.
  • 20% at 5 kb.

Sample Age and Storage Considerations

FFPE block age affects fusion detection success. Older blocks (over 2 years) show higher failure rates. Resection specimens also suffer from RNA degradation due to longer ischemic times.

Pre-analytical Factors

Several pre-analytical factors influence RNA quality:

  • Cold ischemic time should be under 30 minutes for optimal results.
  • Fixation conditions: 10:1 fixative volume using 10% neutral buffered formalin.
  • Fixation duration: 6-48 hours for biopsies.
  • Tissue thickness: Affects fixation quality.

RNA fusion detection is vital in precision cancer medicine. RNA-seq provides superior sensitivity, enabling accurate fusion detection, comprehensive characterization, and functional monitoring. 

Limitations and Ongoing Research Needs for RNA Fusion in Cancer Detection 

Limitations and Ongoing Research Needs for RNA Fusion in Cancer Detection 

RNA fusion detection has advanced precision oncology, but several challenges still exist. Addressing these issues is crucial to maximizing the utility and reliability of RNA-based fusion assays in cancer care. 

1. RNA Quality and Sample Material

Degraded RNA in FFPE samples, older archives, or necrotic tissue leads to poor sequencing and false negatives. Heterogeneous samples with multiple cell types complicate RNA extraction and detection, reducing test accuracy. Pre-analytical factors (such as sample age, processing, and necrosis) greatly affect fusion detection, especially in older paraffin blocks.

2. Expression-Dependent Detection

RNA-seq detects expressed fusion transcripts. However, it may miss fusions that are low in expression or found in silenced genes, leaving out clinically relevant drivers. RNA-based methods often fail to capture all the alterations identified by DNA sequencing. To maximize sensitivity, it’s necessary to combine DNA and RNA sequencing.

3. Technical and Bioinformatics Limitations

The lack of standardized quality control across fusion assays results in inconsistent outcomes. Sequencing artifacts and low transcript abundance can cause misalignment, reducing the accuracy of novel fusion calls. Rare or novel fusion partners are difficult to interpret and often require confirmation with alternative methods.

4. Panel and Assay Coverage

RT-PCR and targeted panels can only detect known fusion genes. Rare fusions may go undetected unless whole-transcriptome sequencing is used, but this is more expensive and less sensitive for low-expressed fusions. Single-gene assays cannot provide broad clinical coverage and fail to assess assay robustness.

5. Cost-Effectiveness and Turnaround Time

Comprehensive fusion detection requires both DNA and RNA sequencing, which increases costs and turnaround times. While parallel approaches maximize detection, they risk delays and may miss some fusions.

However, AI-driven solutions are emerging to overcome these obstacles. There is a pressing need for standardized protocols, better sample processing, and more accessible sequencing platforms.

Overcome RNA Fusion Detection Challenges With Biostate AI

Biostate AI provides a complete RNA sequencing solution that addresses these challenges. From sample collection to final insights, it offers precision and accessibility. With total RNA sequencing and AI-driven analytics, Biostate AI helps researchers focus on their work without dealing with complex lab procedures. The platform delivers high-quality results at an affordable price.

Key Features:

  • Unbeatable Pricing: Achieve high-quality sequencing results starting at $80/sample, making comprehensive fusion detection accessible to more research teams.
  • Rapid Turnaround: Receive results in just 1–3 weeks, significantly reducing delays in critical cancer diagnostic workflows.
  • Complete Transcriptome Insights: Benefit from comprehensive RNA-Seq covering both mRNA and non-coding RNA, ensuring no fusion events are missed due to limited panel coverage.
  • AI-Driven Analysis: Access powerful, intuitive insights with OmicsWeb AI, removing bioinformatics barriers and standardizing result interpretation.
  • Minimal Sample Requirement: Process samples as small as 10µL blood, 10ng RNA, or 1 FFPE slide, maximizing utility from precious clinical specimens.
  • Low RIN Compatibility: Work with RNA samples having RIN as low as 2 (compared to typical ≥5 requirements), addressing the challenge of degraded FFPE samples.
  • Multi-omics Integration: Seamlessly combine RNA-Seq with WGS and methylation data through the OmicsWeb platform, providing a comprehensive approach to maximize fusion detection sensitivity.
  • Disease Prediction AI: Turn RNA fusion data into accurate disease predictions with Biobase, achieving 89% accuracy in drug toxicity prediction and 70% accuracy in therapy selection.

This integrated approach of Biostate AI eliminates the fragmented workflows and technical barriers that have historically limited RNA fusion detection. It democratizes access to advanced molecular diagnostics while maintaining the highest standards of accuracy and reliability.

Final Words!

RNA fusion in cancer detection offers unprecedented insights into tumor biology, enabling precision medicine approaches. From identifying driver mutations to guiding targeted therapies, RNA fusions serve as powerful biomarkers that transform how we detect, diagnose, and treat cancer.

Biostate AI eliminates traditional barriers to RNA fusion analysis with comprehensive sequencing solutions starting at just $80 per sample. Our AI-powered platform handles degraded FFPE samples, delivers results in 1-3 weeks, and provides intuitive analytics, making advanced molecular diagnostics accessible and affordable for researchers worldwide.

Ready to advance your cancer research with cutting-edge RNA fusion detection? Get your quote today and discover how Biostate AI can accelerate your path to breakthrough discoveries.

FAQs

1. Does insurance cover RNA fusion testing for cancer?

Most private insurance companies and Medicare cover RNA fusion testing when a doctor medically indicates it, though coverage policies vary between providers. Most patients typically pay $0-$250 out of pocket, though costs without insurance can range from $100 to $2,000. It’s recommended to contact your insurance provider before testing to understand your specific coverage and any pre-authorization requirements.

2. How long does RNA fusion testing take to get results?

While traditional methods like RT-PCR can provide results within days, comprehensive RNA sequencing for fusion detection typically takes 1-3 weeks from sample submission to final report. The timeline depends on factors such as sample quality, the complexity of analysis required, and laboratory workload. Emergency or urgent cases may have expedited processing options available.

3. Can RNA fusion testing be done on old biopsy samples?

Yes, RNA fusion testing can be performed on archived formalin-fixed paraffin-embedded (FFPE) tissue samples, even those several years old. However, older samples may have degraded RNA quality, which can affect test sensitivity. Poor quality RNA decreases the likelihood of detecting fusion transcripts, particularly for low-expression fusions. Modern testing platforms are increasingly compatible with degraded samples, with some accepting RNA with integrity numbers as low as 2.

4. What happens if my RNA fusion test comes back negative?

A negative RNA fusion test doesn’t rule out cancer or the presence of other genetic alterations. It simply means no detectable fusion genes were found in the analyzed sample. Your oncologist may recommend additional testing, such as DNA sequencing, immunohistochemistry, or other molecular tests to identify alternative therapeutic targets. The treatment plan will be based on the complete clinical picture, including tumor type, staging, and other available biomarkers.

Leave a Comment

Your email address will not be published. Required fields are marked *