Evaluating the Effectiveness of Artificial Intelligence in Predicting Adverse Drug Reactions among Cancer Patients: A Systematic Review and Meta-Analysis

Read original: arXiv:2404.05762 - Published 4/10/2024 by Fatma Zahra Abdeldjouad, Menaouer Brahami, Mohammed Sabri
Total Score

0

🤯

Sign in to get full access

or

If you already have an account, we'll log you in

Overview

  • Adverse drug reactions (ADRs) significantly impact patient outcomes and healthcare costs in cancer treatment
  • Using artificial intelligence (AI) to predict ADRs in real-time could revolutionize oncology care
  • This study aims to assess the performance of AI models in predicting ADRs in cancer patients
  • This is the first systematic review and meta-analysis on this topic

Plain English Explanation

Cancer treatments can sometimes cause unwanted side effects, known as adverse drug reactions (ADRs). These ADRs can negatively impact a patient's health and increase the cost of their care. Leveraging AI and machine learning models to predict ADRs in cancer patients in real-time could be a game-changer, allowing doctors to better manage these issues and improve patient outcomes.

This study reviewed the existing research on using AI to predict ADRs in cancer patients. The researchers searched various scientific databases for relevant studies published between 2018 and 2023. They identified 17 studies that met their criteria, which included using AI algorithms like machine learning and deep learning to predict specific ADRs like heart, liver, kidney, and blood cell problems.

The researchers analyzed the results of these 17 studies and found that the AI models demonstrated high sensitivity (ability to correctly identify patients who will experience an ADR) and specificity (ability to correctly identify patients who won't experience an ADR). The models also had a strong overall performance, as measured by the area under the curve (AUC) metric.

Interestingly, the study found that biomarkers - measurable indicators of some medical state or condition - were effective in predicting ADRs, but were only used in about half of the reviewed studies. This suggests there is room for improvement in incorporating diverse data sources to enhance the accuracy of these AI-powered ADR prediction models.

Technical Explanation

The researchers conducted a systematic review and meta-analysis to assess the performance of AI models in predicting adverse drug reactions (ADRs) in cancer patients. They searched Scopus, PubMed, IEEE Xplore, and ACM Digital Library databases for relevant studies published in English, French, and Arabic between January 1, 2018, and August 20, 2023.

The inclusion criteria were: (1) peer-reviewed research articles; (2) use of AI algorithms (machine learning, deep learning, knowledge graphs); (3) studies aimed to predict specific ADRs (cardiotoxicity, neutropenia, nephrotoxicity, hepatotoxicity); and (4) studies focused on cancer patients.

Out of 332 screened articles, 17 studies (5%) involving 93,248 oncology patients from 17 countries were included in the systematic review. Ten of these studies were further synthesized in the meta-analysis. The researchers used a random-effects model to pool the sensitivity, specificity, and AUC of the included studies.

The pooled results showed that the ADR predictive models had a sensitivity of 0.82 (95% CI: 0.69, 0.9), a specificity of 0.84 (95% CI: 0.75, 0.9), and an AUC of 0.83 (95% CI: 0.77, 0.87). This indicates that the AI models demonstrated high accuracy in predicting ADRs in cancer patients.

The study also found that biomarkers, which were used in only about half of the reviewed studies, proved effective in predicting ADRs. This suggests that incorporating diverse data sources could further improve the performance of these AI-powered ADR prediction models.

Critical Analysis

The systematic review and meta-analysis provide valuable insights into the performance of AI models in predicting adverse drug reactions (ADRs) in cancer patients. The researchers followed a rigorous methodology, including a comprehensive literature search and a thorough evaluation of study quality by multiple reviewers.

However, the study does acknowledge the need for standardized research and multicenter studies to improve the quality of evidence. The heterogeneity of the included studies and the relatively small number of studies (17) may limit the generalizability of the findings.

Additionally, the study focused on specific types of ADRs, such as cardiotoxicity, neutropenia, nephrotoxicity, and hepatotoxicity. While these are important ADRs in cancer treatment, there may be other types of ADRs that were not addressed in this review.

It would also be beneficial to explore the integration of these AI-powered ADR prediction models into clinical decision-making processes and their real-world impact on patient outcomes and healthcare costs. Bridging the gap between data-driven insights and clinical expertise will be crucial for the successful implementation of these technologies in oncology practice.

Conclusion

This systematic review and meta-analysis demonstrate the promising potential of using AI models to predict adverse drug reactions in cancer patients. The high sensitivity, specificity, and overall performance of the reviewed models suggest that this technology could revolutionize oncology care by enabling proactive management of ADRs and improving patient outcomes.

However, further research is needed to standardize the methodologies, expand the scope of ADRs studied, and explore the real-world integration of these AI-powered tools into clinical practice. Continued efforts in this direction could empower biomedical discovery and enhance cancer patient care by bridging the gap between data-driven insights and clinical expertise.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Follow @aimodelsfyi on 𝕏 →

Related Papers

🤯

Total Score

0

Evaluating the Effectiveness of Artificial Intelligence in Predicting Adverse Drug Reactions among Cancer Patients: A Systematic Review and Meta-Analysis

Fatma Zahra Abdeldjouad, Menaouer Brahami, Mohammed Sabri

Adverse drug reactions considerably impact patient outcomes and healthcare costs in cancer therapy. Using artificial intelligence to predict adverse drug reactions in real time could revolutionize oncology treatment. This study aims to assess the performance of artificial intelligence models in predicting adverse drug reactions in patients with cancer. This is the first systematic review and meta-analysis. Scopus, PubMed, IEEE Xplore, and ACM Digital Library databases were searched for studies in English, French, and Arabic from January 1, 2018, to August 20, 2023. The inclusion criteria were: (1) peer-reviewed research articles; (2) use of artificial intelligence algorithms (machine learning, deep learning, knowledge graphs); (3) study aimed to predict adverse drug reactions (cardiotoxicity, neutropenia, nephrotoxicity, hepatotoxicity); (4) study was on cancer patients. The data were extracted and evaluated by three reviewers for study quality. Of the 332 screened articles, 17 studies (5%) involving 93,248 oncology patients from 17 countries were included in the systematic review, of which ten studies synthesized the meta-analysis. A random-effects model was created to pool the sensitivity, specificity, and AUC of the included studies. The pooled results were 0.82 (95% CI:0.69, 0.9), 0.84 (95% CI:0.75, 0.9), and 0.83 (95% CI:0.77, 0.87) for sensitivity, specificity, and AUC, respectively, of ADR predictive models. Biomarkers proved their effectiveness in predicting ADRs, yet they were adopted by only half of the reviewed studies. The use of AI in cancer treatment shows great potential, with models demonstrating high specificity and sensitivity in predicting ADRs. However, standardized research and multicenter studies are needed to improve the quality of evidence. AI can enhance cancer patient care by bridging the gap between data-driven insights and clinical expertise.

Read more

4/10/2024

👀

Total Score

0

Evaluating Physician-AI Interaction for Cancer Management: Paving the Path towards Precision Oncology

Zeshan Hussain, Barbara D. Lam, Fernando A. Acosta-Perez, Irbaz Bin Riaz, Maia Jacobs, Andrew J. Yee, David Sontag

We evaluated how clinicians approach clinical decision-making when given findings from both randomized controlled trials (RCTs) and machine learning (ML) models. To do so, we designed a clinical decision support system (CDSS) that displays survival curves and adverse event information from a synthetic RCT and ML model for 12 patients with multiple myeloma. We conducted an interventional study in a simulated setting to evaluate how clinicians synthesized the available data to make treatment decisions. Participants were invited to participate in a follow-up interview to discuss their choices in an open-ended format. When ML model results were concordant with RCT results, physicians had increased confidence in treatment choice compared to when they were given RCT results alone. When ML model results were discordant with RCT results, the majority of physicians followed the ML model recommendation in their treatment selection. Perceived reliability of the ML model was consistently higher after physicians were provided with data on how it was trained and validated. Follow-up interviews revealed four major themes: (1) variability in what variables participants used for decision-making, (2) perceived advantages to an ML model over RCT data, (3) uncertainty around decision-making when the ML model quality was poor, and (4) perception that this type of study is an important thought exercise for clinicians. Overall, ML-based CDSSs have the potential to change treatment decisions in cancer management. However, meticulous development and validation of these systems as well as clinician training are required before deployment.

Read more

4/24/2024

Autonomous Artificial Intelligence Agents for Clinical Decision Making in Oncology
Total Score

0

Autonomous Artificial Intelligence Agents for Clinical Decision Making in Oncology

Dyke Ferber, Omar S. M. El Nahhas, Georg Wolflein, Isabella C. Wiest, Jan Clusmann, Marie-Elisabeth Le{ss}man, Sebastian Foersch, Jacqueline Lammert, Maximilian Tschochohei, Dirk Jager, Manuel Salto-Tellez, Nikolaus Schultz, Daniel Truhn, Jakob Nikolas Kather

Multimodal artificial intelligence (AI) systems have the potential to enhance clinical decision-making by interpreting various types of medical data. However, the effectiveness of these models across all medical fields is uncertain. Each discipline presents unique challenges that need to be addressed for optimal performance. This complexity is further increased when attempting to integrate different fields into a single model. Here, we introduce an alternative approach to multimodal medical AI that utilizes the generalist capabilities of a large language model (LLM) as a central reasoning engine. This engine autonomously coordinates and deploys a set of specialized medical AI tools. These tools include text, radiology and histopathology image interpretation, genomic data processing, web searches, and document retrieval from medical guidelines. We validate our system across a series of clinical oncology scenarios that closely resemble typical patient care workflows. We show that the system has a high capability in employing appropriate tools (97%), drawing correct conclusions (93.6%), and providing complete (94%), and helpful (89.2%) recommendations for individual patient cases while consistently referencing relevant literature (82.5%) upon instruction. This work provides evidence that LLMs can effectively plan and execute domain-specific models to retrieve or synthesize new information when used as autonomous agents. This enables them to function as specialist, patient-tailored clinical assistants. It also simplifies regulatory compliance by allowing each component tool to be individually validated and approved. We believe, that our work can serve as a proof-of-concept for more advanced LLM-agents in the medical domain.

Read more

4/9/2024

📊

Total Score

0

Coalitions of AI-based Methods Predict 15-Year Risks of Breast Cancer Metastasis Using Real-World Clinical Data with AUC up to 0.9

Xia Jiang, Yijun Zhou, Alan Wells, Adam Brufsky

Breast cancer is one of the two cancers responsible for the most deaths in women, with about 42,000 deaths each year in the US. That there are over 300,000 breast cancers newly diagnosed each year suggests that only a fraction of the cancers result in mortality. Thus, most of the women undergo seemingly curative treatment for localized cancers, but a significant later succumb to metastatic disease for which current treatments are only temporizing for the vast majority. The current prognostic metrics are of little actionable value for 4 of the 5 women seemingly cured after local treatment, and many women are exposed to morbid and even mortal adjuvant therapies unnecessarily, with these adjuvant therapies reducing metastatic recurrence by only a third. Thus, there is a need for better prognostics to target aggressive treatment at those who are likely to relapse and spare those who were actually cured. While there is a plethora of molecular and tumor-marker assays in use and under-development to detect recurrence early, these are time consuming, expensive and still often un-validated as to actionable prognostic utility. A different approach would use large data techniques to determine clinical and histopathological parameters that would provide accurate prognostics using existing data. Herein, we report on machine learning, together with grid search and Bayesian Networks to develop algorithms that present a AUC of up to 0.9 in ROC analyses, using only extant data. Such algorithms could be rapidly translated to clinical management as they do not require testing beyond routine tumor evaluations.

Read more

8/30/2024