top of page
Search
  • Writer's pictureSandy Sanbar

AI Medical Devices and Algorithms in Pathology and Genomics

FDA-Approved AI Medical Devices and Algorithms in pathology and for pathologists are tools designed to assist in the diagnosis and management of various diseases by analyzing digital pathology slides, genomic data, and other relevant information. These tools aim to augment the expertise of pathologists, ultimately leading to more accurate diagnoses and better patient care.

 

Landscape of FDA-approved AI Medical Devices and Algorithms


This is a rapidly evolving area.  New devices and algorithms are being developed and approved regularly.  Several AI medical devices and algorithms in the field of pathology are either in the FDA approval process or are expected to seek approval in the near future.

 

AI has shown significant promise in improving diagnostic accuracy in pathology by assisting pathologists in interpreting complex tissue samples and cellular images. Here are some examples of how AI is enhancing diagnostic accuracy in this field:

 

  1. Cancer Diagnosis and Classification: AI algorithms, particularly those using deep learning, have been trained to recognize the patterns of various types of cancer in digital pathology slides. For instance, AI can help in distinguishing between benign and malignant tumors, identifying the subtype of cancer, and even predicting patient outcomes based on the analysis of tissue samples.

  2. Detection of Genetic Alterations: AI can analyze genetic information from pathology samples to identify mutations or alterations that may be relevant for diagnosis or treatment. This is particularly useful in cancer pathology, where genetic mutations can inform targeted therapy options.

  3. Immune Cell Profiling: AI tools can assess the immune cell composition within a tumor, which is important for understanding the tumor microenvironment and the patient's immune response. This information can be crucial for predicting the effectiveness of immunotherapies.

  4. Quantitative Analysis: AI can perform quantitative analyses of pathology images, providing precise measurements of cellular features that may be difficult for human pathologists to assess consistently. This can include counting cells, measuring the area of tissues, and quantifying the expression of proteins.

  5. Quality Control: AI systems can help in ensuring the quality of pathology slides by automatically detecting issues such as poor staining or tissue artifacts that could affect diagnostic accuracy.

  6. Real-time Diagnostic Support: During live examinations, AI can provide real-time assistance to pathologists, highlighting areas of concern and suggesting potential diagnoses, which can speed up the diagnostic process and improve accuracy.

  7. Predictive Analytics: AI can analyze pathology data to predict patient outcomes, such as the likelihood of cancer recurrence or the response to specific treatments. This can help in making more informed treatment decisions.

  8. Integration with Other Data Types: AI can integrate pathology data with other types of medical data, such as radiology images, lab results, and electronic health records, to provide a more comprehensive diagnostic assessment.


Companies and research institutions are developing AI-driven pathology tools that are being tested in clinical settings. Examples include Paige.AI, which offers AI-based software for prostate and breast cancer diagnosis, and the use of Google's deep learning algorithms to detect metastatic breast cancer in lymph node biopsy images.


 

AI Solutions that could potentially receive FDA approval


Companies and research institutions are actively working on AI solutions that could potentially receive FDA approval in the coming years. These solutions may include:

 

  1. Advanced image analysis tools for digital pathology that can detect and classify various types of cancer with high accuracy.

  2. AI systems for predicting patient outcomes based on pathology reports and genetic data.

  3. Algorithms for optimizing treatment plans by integrating pathology findings with other clinical data.

  4. AI-driven quality control tools for pathology labs to ensure the accuracy and consistency of diagnoses.

  5. Machine learning models for the early detection of diseases from tissue samples.

 

As of May 2024, the FDA has approved over 870 AI medical devices and algorithms that have undergone clinical trials and studies to support their efficacy and safety before receiving regulatory clearance. These studies are typically designed to demonstrate that the AI tool can perform its intended function with a level of accuracy and reliability that is comparable to or better than traditional methods or human experts.

 

Devices and algorithms in the field of pathology include:

 

  1. Paige.AI has products that can detect  cancer in prostate and breast tissue samples.  It offers AI algorithms for digital pathology, including tools for prostate cancer diagnosis and breast cancer tumor detection. These algorithms help pathologists by providing additional insights derived from the analysis of whole slide images.  For AI tools like Paige.AI, clinical validation includes demonstrating the algorithm's ability to detect and classify cancer cells in pathology slides accurately. Studies would compare the AI's performance with that of experienced pathologists.

  2. Tempus xT is an artificial intelligence-driven clinical laboratory test that analyzes DNA sequencing data from a patient's tumor and whole blood sample. It is used to help oncologists make informed treatment decisions for patients with certain types of cancer.   It is osed for the genomic sequencing analysis for cancer treatment decisions.  Clinical trials for Tempus xT would involve analyzing the genomic data of cancer patients and showing that the insights provided by the AI lead to more effective treatment plans and improved patient outcomes.

  3. IBM Watson for Oncology: IBM Watson for Oncology is an AI system that assists oncologists in making treatment recommendations for cancer patients. It analyzes patient data and medical literature to provide evidence-based treatment options.  IBM Watson for Oncology has been evaluated in various clinical settings to ensure it provides recommendations that are consistent with those made by oncologists. Studies would assess the system's ability to analyze complex medical data and provide useful treatment options.

  4. IDx-DR is an AI-based diagnostic device that is used for the automated detectioin and diagnosis of diabetic retinopathy and diabetic macular edema in retinal images.  It is intended for use by healthcare providers to screen for diabetic retinopathy and diabetic macular edema in adults diagnosed with diabetes without signs of diabetic retinopathy.  The clinical validation of IDx-DR involved multiple studies that compared its performance in detecting diabetic retinopathy against the gold standard of manual grading by trained professionals. These studies showed that the AI system could accurately identify patients with diabetic retinopathy and diabetic macular edema which supported its approval for use in screening.

  5. Viz.AI Contact is an AI-powered software platform designed to facilitate faster stroke care.  It is an AI to analyze CT scans and notify stroke specialists of potential large vessel occlusions, which can help in the rapid identification and treatment of stroke patients. It is used for the detection of large vessel occlusions in stroke patients using CT scans.  Clinical studies for Viz.AI Contact would involve assessing the software's ability to accurately identify large vessel occlusions in stroke patients and demonstrating that its use leads to faster treatment times and improved patient outcomes.

  6. Arterys Cardio AI is an AI software is used for the analysis of cardiac MRI images. It provides automated quantification of cardiac function and tissue characterization, aiding in the diagnosis and management of cardiovascular diseases.  The efficacy of Arterys Cardio AI would be supported by studies comparing its automated analysis of cardiac MRI images with manual analysis by cardiologists. These studies would evaluate the software's accuracy in quantifying cardiac function and tissue characterization.

  7. Aidoc provides multiple AI algorithms for radiology, including those that help in the detection of acute conditions such as intracranial hemorrhage, stroke, and pulmonary embolism. These algorithms analyze CT scans to help radiologists prioritize and expedite the diagnosis of time-sensitive conditions.  Aidoc's algorithms have been evaluated in clinical settings to ensure they can effectively assist radiologists in detecting acute conditions like intracranial hemorrhage and stroke. Studies often focus on the algorithm's sensitivity, specificity, and its impact on patient outcomes and workflow efficiency.

  8. Freenome is an AI-based blood test is designed to detect early-stage cancer using a combination of AI and epigenetic biomarkers. The test is still under development but has shown promise in early detection research.

  9. EchoNous EchoWise is an AI-powered handheld ultrasound system that provides automated measurements and analysis, enabling point-of-care ultrasound imaging for a variety of clinical applications.  Clinical trials for EchoNous EchoWise would focus on the device's ability to provide accurate ultrasound imaging and automated measurements, comparing its performance with traditional ultrasound systems.

  10. GeneEx is an AI-driven software used for the analysis of gene expression data in breast cancer patients. It helps in the classification of breast cancer subtypes and can be used to guide treatment decisions.  Studies supporting GeneEx would involve demonstrating the software's accuracy in classifying breast cancer subtypes based on gene expression data and its utility in guiding treatment decisions.


The specific studies and clinical trials for each AI medical device can be found in medical journals, FDA documentation, and the manufacturers' websites.

 

Accuracy of Traditional versus AI Methods when Detecting Genetic Alterations


AI has the potential to surpass or significantly complement traditional methods in terms of accuracy when detecting genetic alterations in pathology samples. Here are some key points comparing AI to traditional methods:

 

  1. High Throughput: AI can analyze large volumes of genetic data much faster than traditional methods, allowing for the detection of genetic alterations at scale.

  2. Consistency: AI algorithms apply the same criteria consistently across all samples, reducing the variability that can occur with manual analysis by human pathologists.

  3. Sensitivity and Specificity: AI can be trained to achieve high sensitivity (ability to detect true positives) and specificity (ability to detect true negatives). Once trained on large, high-quality datasets, AI can often match or exceed the performance of traditional methods.

  4. Complex Pattern Recognition: AI, particularly deep learning models, excels at recognizing complex patterns in data. This capability is particularly useful for detecting subtle genetic alterations that may be missed by traditional methods.

  5. Integration of Multiple Data Types: AI can integrate various types of data, including genetic sequences, gene expression profiles, and clinical information, to provide a more comprehensive analysis of genetic alterations.

  6. Learning from New Data: AI systems can continuously learn from new data, improving their accuracy over time as they are exposed to a wider range of genetic alterations and clinical outcomes.

  7. Cost-Effectiveness: Once implemented, AI-driven analysis can be more cost-effective than some traditional methods, especially for high-throughput analysis.


However, it's important to note that the performance of AI in detecting genetic alterations depends on several factors:

 

  • Quality of Training Data: The accuracy of AI is heavily dependent on the quality and representativeness of the data it was trained on. Biases or limitations in the training data can affect the AI's performance.

  • Algorithm Design: The design of the AI algorithm, including the choice of model and features, will impact its ability to accurately detect genetic alterations.

  • Validation and Testing: Rigorous validation and testing against gold standard methods are necessary to ensure that AI tools perform accurately across different types of samples and clinical scenarios.

  • Regulatory Approval: AI tools must undergo regulatory scrutiny to ensure they meet the required standards for clinical use.


Thus, AI has the potential to match or exceed traditional methods in accuracy for detecting genetic alterations, particularly when it comes to speed, consistency, and the ability to handle large and complex datasets. However, the success of AI in this context depends on careful development, validation, and ongoing improvement of the algorithms used.

 

AI Tools for Detecting Genetic Alterations

 

AI tools for detecting genetic alterations are becoming increasingly sophisticated and are being used in both research and clinical settings. Here are some examples of AI tools and platforms that are currently used or in development for this purpose:

 

  1. Google's DeepVariant: Developed by Google's AI team, DeepVariant is an open-source deep learning-based variant caller that uses a deep neural network to analyze next-generation sequencing (NGS) data and identify genetic variants with high accuracy.

  2. Illumina's DRAGEN (Dynamic Read Analysis for GENomics): DRAGEN is a bioinformatics platform that uses AI and machine learning to accelerate the analysis of genetic data. It can rapidly align, sort, and call variants from NGS data, enabling faster detection of genetic alterations.

  3. Fabric Genomics' Genome Interpretation Network (GIN): GIN is an AI-driven platform that helps clinical labs and researchers interpret genetic data. It uses natural language processing and machine learning to analyze genetic variants and provide insights into their clinical significance.

  4. Tempus' xT Test: Tempus is a healthcare technology company that uses AI to analyze genetic data from cancer patients. Its xT test analyzes DNA sequencing data from a patient's tumor and whole blood sample to identify genetic alterations that can inform treatment decisions.

  5. Foundation Medicine's FoundationOne and FoundationOne Liquid: These are comprehensive genomic profiling assays that use AI to analyze tumor tissue and circulating tumor DNA (ctDNA), respectively. They identify genetic alterations to help oncologists select the most effective treatment options for cancer patients.

  6. Guardant Health's Guardant360: This is a blood test that uses AI to analyze ctDNA for early detection of cancer and to identify genetic alterations that can guide treatment decisions for advanced cancer patients.

  7. Grail's Galleri test: Grail is developing an AI-powered multi-cancer early detection blood test that aims to detect more than 50 types of cancer through the analysis of cell-free DNA (cfDNA) and other biomarkers.

  8. NantHealth's GPS Cancer Test: This AI-driven test analyzes the molecular and genetic information of a patient's tumor to provide personalized treatment recommendations based on the detection of genetic alterations.

These tools and tests leverage AI to enhance the accuracy and efficiency of genetic alteration detection, which is crucial for precision medicine and the development of targeted therapies. As AI technology continues to evolve, we can expect to see more advanced tools being developed and integrated into clinical practice.

 

Fabric Genomics' Genome Interpretation Network (GIN)

 

This AI algorithm offers several advantages for clinical labs that are involved in genetic testing and interpretation. Here are some of the main advantages:

 

  1. Improved Efficiency: GIN can automate many steps in the genetic interpretation process, which can significantly reduce the time and effort required by lab personnel. This can lead to faster turnaround times for test results, allowing for more timely clinical decisions.

  2. Enhanced Accuracy: The use of AI and machine learning in GIN helps to standardize the interpretation of genetic variants, reducing the risk of human error and potentially improving the accuracy of diagnoses.

  3. Comprehensive Analysis: GIN can analyze a wide range of genetic data, including whole exome and whole genome sequencing. It integrates information from various databases and literature to provide a comprehensive understanding of the clinical significance of genetic variants.

  4. Scalability: As a software platform, GIN can scale to handle large volumes of genetic data, making it suitable for labs with high-throughput needs or those that expect growth in genetic testing volumes.

  5. Regulatory Compliance: GIN is designed to comply with regulatory standards, such as those set by the College of American Pathologists (CAP) and the Clinical Laboratory Improvement Amendments (CLIA). This can help labs ensure that their genetic testing and interpretation practices meet the required standards.

  6. Integration with Electronic Health Records (EHRs): GIN can integrate with EHRs, allowing for a more seamless flow of genetic information into the clinical workflow. This can improve the accessibility of genetic test results and their integration into patient care.

  7. Continuous Learning: The AI component of GIN can continuously learn from new data and updates to genetic databases, ensuring that the interpretations provided are based on the latest scientific evidence and knowledge.

  8. Support for Variant Reassessment: GIN can help labs in the reassessment of genetic variants as new evidence emerges, which is important for maintaining the accuracy of genetic diagnoses over time.

  9. Customization and Flexibility: Labs can customize GIN to fit their specific workflows and requirements, and the platform can be flexible enough to adapt to changing needs or new testing methodologies.

  10. Expert Support: Fabric Genomics may provide expert support and training to help labs maximize the use of GIN, which can be particularly valuable for labs new to genetic testing or those looking to expand their services.


By leveraging AI and machine learning, Fabric Genomics' Genome Interpretation Network aims to provide clinical labs with a powerful tool that enhances the quality, speed, and efficiency of genetic interpretation, ultimately contributing to better patient care through more precise diagnostics.


AI-driven quality control tools for pathology labs


These tools are designed to ensure the accuracy and consistency of diagnoses by automating the review of pathology slides and related data. They use machine learning algorithms to analyze digital pathology images and other relevant information to identify potential errors or inconsistencies.


Function of AI-driven quality control tools for pathology include:


  1. Image Analysis: AI algorithms are trained to recognize patterns and features in digital pathology slides that are indicative of specific diseases or conditions. They can detect cellular structures, tissue arrangements, and other morphological characteristics with high precision.

  2. Consistency Checks: The AI system can compare current diagnoses with previous ones made by the same lab or with established diagnostic criteria. This helps in maintaining consistency in the interpretation of slides and reduces the likelihood of discrepancies.

  3. Error Detection: AI can flag slides that may contain errors or require a second look. This could be due to issues such as poor slide preparation, staining artifacts, or areas of ambiguity that are difficult to interpret.

  4. Workflow Integration: Quality control tools are integrated into the lab's workflow, allowing for real-time or near-real-time analysis of slides as they are being processed. This immediate feedback can help pathologists address any issues promptly.

  5. Continuous Learning: Some AI systems are designed to learn from the feedback provided by pathologists. When a pathologist confirms or corrects a diagnosis, the AI algorithm can use this information to improve its future analyses, making the quality control process more accurate over time.

  6. Data Management: These tools can also help in managing and organizing the vast amounts of data generated in pathology labs. They can track the history of slide reviews, diagnostic decisions, and any adjustments made, which is crucial for maintaining high-quality standards and for auditing purposes.

  7. Regulatory Compliance: AI-driven quality control tools can assist labs in complying with regulatory requirements by providing documentation of the review process and ensuring that all diagnoses meet the necessary standards.

  8. Education and Training: These systems can be used to train new pathologists by providing examples of correct diagnoses and highlighting common pitfalls. They can also serve as a reference for experienced pathologists when dealing with complex or rare cases.


The implementation of AI-driven quality control tools in pathology labs can significantly enhance the reliability of diagnoses, reduce the rate of false negatives and positives, and ultimately improve patient care.

7 views0 comments

Comments


bottom of page