• Skip to primary navigation
  • Skip to main content
  • Skip to primary sidebar

Pharmacovigilance Analytics

Your best resource for PV analytics news, content and innovation!

  • Home
  • Pharmacovigilance Analytics
    • Sources of data
    • Methods and tools
  • Signal Detection and Management in Pharmacovigilance
  • News
  • Opinion
  • About
  • Glossary

artificial intelligence

Predictive Models for Identifying Drug Risks: Enhancing Pharmaceutical Safety

November 28, 2023 by Jose Rossello 1 Comment

In the field of healthcare, leveraging the power of predictive models to identify drug risks before they affect patients is becoming increasingly crucial. With many medications on the market and more entering it each year, the ability to predict adverse drug reactions can significantly enhance patient safety and therapeutic effectiveness. These models are built on a variety of statistical and machine learning techniques that can analyze extensive datasets to forecast potential drug risks.

This predictive capability is particularly important in the context of personalized medicine, where a patient’s unique characteristics, such as genetic makeup, can affect their response to drugs. Integrating diverse biomedical data sources, including electronic health records and genomic data, helps create more accurate and individualized risk profiles. As machine learning algorithms grow more sophisticated, they open new avenues in drug risk assessment, allowing researchers to uncover complex patterns and interactions that may not be apparent using traditional statistical methods.

Key Takeaways

  • Predictive models facilitate early detection of drug risks, improving patient outcomes.
  • Machine learning techniques enhance the precision of drug risk assessments.
  • Integration of diverse data sources is pivotal for advancing personalized medicine.

Fundamentals of Predictive Modeling

Predictive modeling is a cornerstone in the realm of drug development, leveraging the power of both statistical analysis and machine learning techniques to anticipate drug risks.

Overview of Predictive Models

Predictive models are computational tools that project future events based on historical data. In the context of drug development, these models analyze patterns in clinical trial data to forecast potential adverse drug events. The efficacy of these models largely hinges on the data quality and the appropriateness of the statistical or machine learning techniques employed.

Importance in Drug Development

Within drug development, the application of predictive analytics is crucial for identifying FDA-approved drugs’ side effects that might not be evident during initial clinical trials. By efficiently signaling potential risks, predictive models can save pharmaceutical companies time and resources, while protecting patient safety.

Key Statistical and Machine Learning Techniques

Key techniques in predictive modeling encompass a wide range of statistical and machine learning approaches. Statistical analysis forms the basis for understanding data relationships, while machine learning techniques like natural language processing (NLP) and next-generation sequencing (NGS) parse through complex, unstructured data sets to identify drug risk. Within this landscape, feature selection is imperative to refine models, focusing on the most relevant predictors, such as patient phenotypes and genetic profiles acquired from high-throughput screening.

Challenges and Considerations

The accuracy of predictive models is contingent upon the volume and quality of the data. Factors like missing information, inaccuracies, and biased data sets can lead to underperforming models. Furthermore, while statistical models rely on pre-determined equations, machine learning models may require vast amounts of training data to learn and adapt. It is also crucial to address the interpretability of the model to ensure that the predictions can be understood and trusted by medical professionals.

Advancements in Computational Tools

The integration of advanced computational models has propelled the field forward. Developments in artificial intelligence, especially in areas such as natural language processing and machine learning, allow for the synthesis of large datasets from diverse sources like electronic health records and literature databases. This multi-faceted approach enriches predictive modeling by introducing broader context and facilitating deeper insights into drug safety and efficacy.

Machine Learning in Drug Risk Assessment

Machine learning plays a crucial role in enhancing drug risk assessment by providing sophisticated models that can predict drug response, identify potential risks, and streamline drug development.

Predictive Algorithms for Drug Development

Researchers utilize predictive algorithms for drug development to reduce risks and costs associated with clinical trials. These algorithms analyze vast datasets to forecast adverse reactions and efficacy, enabling a more targeted approach in early-stage research.

Machine Learning Approaches to Oncology

In the realm of precision oncology, machine learning approaches refine drug sensitivity prediction by incorporating global cancer statistics and omic profiles. They focus on identifying biomarkers that signal a tumor’s potential reaction to treatment, thus personalizing therapy for cancer patients.

Predictive Modelling for Precision Medicine

Predictive modelling has shown promise in precision medicine, particularly personalized medicine, where treatments are adapted to the individual’s genetic makeup. By analyzing cell line data and biomarkers, these models suggest optimal treatment strategies for the individual’s specific disease profile.

Multi-Task Learning and Network Approaches

Multi-task learning and complex neural network architectures contribute to a better understanding of drug responses. By learning from diverse but related tasks simultaneously, multi-task frameworks can discern subtle patterns across various types of drugs and diseases, leading to more reliable predictors of drug efficacy and toxicity.

Evaluating Drug Response and Sensitivity

Accurate evaluation of drug response and sensitivity is achieved through advanced machine learning algorithms that process comprehensive datasets. These algorithms enable the exploration of drug repurposing opportunities by identifying potential new uses for existing drugs based on drug response prediction analyses and have become essential tools in the development of targeted therapies.

Integrating Diverse Biomedical Data Sources

Successful predictive modeling in drug risk identification crucially depends on the integration of diverse biomedical data sources ranging from genetic information to clinical data. This complex convergence is geared towards understanding drug risks and patient-specific responses at a granular level.

Application of Omics Data in Predictive Modeling

Omics data, including gene expression and RNA-sequencing, offers a comprehensive view of an organism’s biological processes. Predictive models utilize omics profiles to identify patterns that may indicate adverse drug reactions. For instance, variations in gene expression can suggest how an individual might respond to a given drug, potentially reducing the risk of unanticipated effects.

Incorporating Genetic and Molecular Features

Predictive models often feature genetic and molecular data, like mutation information, to forecast drug efficacy and safety. Deep learning techniques analyze these molecular features in concert with pharmacogenomic data, enhancing the precision with which drug risks are identified. This approach strives to tailor drug administration strategies to individuals’ genetic makeup, thereby mitigating potential risks.

Leveraging Clinical and Pharmacogenomic Data

When it comes to personalizing medicine, incorporating clinical studies and pharmacogenomic interactions into predictive models is key. These data sources are instrumental in understanding how genetic factors influence drug response. Moreover, pharmacogenomic data provides critical insights into the optimal drug choice and dosing for each patient, based on their unique genetic profile.

Use of Real-World Data and Electronic Health Records

Lastly, real-world data derived from electronic health records (EHRs) serves as a rich resource for predictive models. Here, natural language processing (NLP) techniques are used to extract meaningful information from unstructured clinical narratives. This integration of real-world evidence with traditional data facilitates a comprehensive understanding of drug risks in diverse patient populations.

Specialized Predictive Models in Oncology

Precision oncology leverages specialized predictive models to tailor cancer treatment, enhance drug discovery, and mitigate risk. These models integrate various data sets, from genetic biomarkers to clinical trial outcomes, thus refining drug response prediction and aiding in the development of targeted therapies.

Targeting Cancer Treatment Through Predictive Models

Predictive models in oncology are pivotal for the identification of targeted therapies. These models allow researchers to effectively predict how certain cancers will respond to specific drugs, greatly impacting the successful outcomes in clinical trials. For instance, the utilization of machine learning approaches for drug response prediction in cancer has shown promising potential to enhance treatment personalization.

Advances in Precision Oncology and Biomarkers

Precision oncology has transformed cancer treatment through the use of biomarkers. These are biological indicators that help in predicting how a patient will respond to a particular therapy, leading to a more efficient drug discovery and development process. The assessment of biomarkers has become a cornerstone in developing novel therapeutic strategies, including combination therapy.

Innovations in Cancer Risk Prediction Models

Innovations in risk prediction models enable early detection of potential adverse events, including susceptibility to opioid use disorder in pain management post-treatment. These models are integral to understanding the varied risks associated with cancer and its treatment, contributing to more informed decision-making in patient care.

Multi-Omic Correlates in Cancer Therapies

The integration of multi-omic correlates—genes, proteins, and other molecular data—into predictive models has greatly enhanced the understanding of cancer biology. This comprehensive approach improves the prediction of drug efficacy and may inform the design of drug combination prediction models, offering insights into how different therapies can be effectively combined.

Utilizing Patient-Derived Models for Therapy Prediction

Patient-derived xenograft mouse models are increasingly used to anticipate how cancer patients will respond to treatments. These models, which involve transplanting human tumors into mice, provide a more accurate representation of drug response prediction in preclinical settings, helping to identify the most promising treatments for progression into clinical trials.

Predictive Models for Drug Repositioning and Combination Therapies

Predictive models are transforming the field of pharmacology, particularly in the realms of drug repurposing and combination therapies. By leveraging predictive analytics and machine learning techniques, researchers can identify new applications for existing drugs and anticipate the efficacy of drug combinations.

Drug Repurposing Using Predictive Analytics

The process of drug repurposing involves identifying new therapeutic potentials for existing drugs. Predictive analytics apply computational models that integrate diverse datasets to forecast new drug applications. For instance, autoencoders, a form of neural networks, can deduce chemical properties from vast chemical libraries, pinpointing candidates for repurposing with a higher likelihood of success.

Predicting Efficacy of Drug Combinations

The precision of drug combination prediction hinges on comprehending how drugs interact. Predictive models use cell line data and drug sensitivity prediction algorithms to establish which combinations might be most effective. This approach is pivotal for combination therapy design, where the objective is to maximize therapeutic effects while minimizing adverse interactions.

Utilizing High-Throughput Data for Combination Therapy

High-throughput screening generates extensive data that predictive models analyze to determine potential drug combinations. Through processing cell line data and other biological information, researchers can swiftly evaluate thousands of drug interactions to identify promising combination therapies for further investigation.

Role of Machine Learning in Drug Synergy Prediction

Machine learning is indispensable in predicting drug synergies, whereby the combined effect of drugs exceeds the sum of their individual effects. Machine learning algorithms analyze complex datasets to discern patterns and predict interactions, a task impractical for human analysis alone. Thus, machine learning accelerates the discovery of combination therapies that can be tailored to individual patient needs.

Frontiers and Future Directions

The exploration of predictive models in drug safety is rapidly advancing, bringing forth innovative techniques and integrative approaches. This evolution promises to enhance the precision of adverse event predictions and tailor drug safety assessments to individual patient profiles.

Emerging Techniques in Predictive Modeling

The field of predictive modeling is witnessing significant advancements through the adoption of deep learning and variational autoencoders. These techniques are especially proficient in decoding complex, high-dimensional biomedical data, leading to more accurate predictions of drug risks. Researchers are now leveraging self-learning algorithms to refine the identification of potential adverse drug reactions from existing databases.

Integrative Approaches to Drug Risk Prediction

To enhance the predictive power of models, scientists are combining statistical methods with machine learning algorithms. This integrative approach utilizes vast arrays of data from clinical trials and real-world evidence, increasing the reliability of predictions. It allows for a more comprehensive evaluation of FDA-approved drugs, integrating various data sources to anticipate and mitigate potential risks.

Potential of AI in Personalized Drug Safety

The potential for AI to drive personalized medicine in the realm of drug safety is immense. Predictive analytics are shifting towards patient-specific models, where individual genetic profiles and medical histories inform the safety profile of medications. This bespoke approach to medicine aims to minimize adverse events by foreseeing how different patients might react to certain drugs.

Regulatory Considerations and Model Validation

For predictive models to be effectively applied in clinical settings, they must undergo stringent validation processes to meet regulatory standards. Regulatory bodies are actively developing frameworks to evaluate the efficacy of predictive models in drug safety. This involves rigorous testing and validation of the models to ensure accuracy and consistency in adverse drug reaction predictions.

Frequently Asked Questions

Predictive modeling leverages historical data to foresee potential drug risks, thus enhancing patient safety. Each question below delves into aspects critical to understanding and improving predictive models in the realm of drug safety.

How can predictive modeling improve the identification of potential drug risks?

Predictive modeling applies algorithms and statistical techniques to analyze data on drug use and outcomes, enabling the early identification of adverse drug events. This approach can highlight risk factors that may not be evident through traditional analysis.

What types of data are most valuable when creating a predictive model for drug safety?

Data that is comprehensive and high-quality, including electronic health records, clinical trial data, and real-world evidence, is invaluable for creating robust predictive models. Detailed information on drug dosage, patient demographics, and prior health history contribute to a model’s accuracy.

What are the key factors that predictive models consider when assessing drug risks?

They typically consider variables such as patient age, genetics, medical history, polypharmacy, and drug interactions to assess the likelihood of adverse events. The chosen factors depend on the specific drug and the context of its use.

How do predictive models differentiate between correlation and causation in drug risk analysis?

Predictive models utilize statistical methods to identify patterns that suggest causal relationships while controlling for confounding variables. Cross-validation and other machine learning techniques can help to differentiate true causation from mere correlation.

What methodologies are commonly used in the development of predictive models for pharmacovigilance?

Methodologies such as multivariable logistic regression, machine learning algorithms, and cross-validated predictive modeling are employed. These approaches are designed to enhance model generalizability and minimize overfitting.

How effective have predictive models been in reducing adverse drug reactions in real-world settings?

Predictive models, when properly designed and applied, have demonstrated effectiveness in reducing the incidence of adverse drug events by alerting healthcare professionals to potential risks, thereby improving patient safety and outcomes.

Jose Rossello
Jose Rossello

Filed Under: Predictive Analytics Tagged With: artificial intelligence

The Proportional Reporting Ratio According to ChatGPT

November 15, 2023 by Jose Rossello Leave a Comment

With this exercise, I wanted to explore this artificial intelligence tool (ChatGPT) and see what we obtain when asking how to calculate the proportional reporting ratio. The results may surprise you!

ChatGPT 3.5 and PRR Calculation

First, let’s try ChatGPT 3.5 (free version). ‘You’, displays my question, and ‘ChatGPT’ displays their response. ‘Editor’s Note’ indicates my comments on the ChatGPT outputs.

You

I would like to know how to calculate the proportional reporting ratio (PRR)

ChatGPT

The Proportional Reporting Ratio (PRR) is a statistical measure used in pharmacovigilance to assess whether a particular adverse event (side effect) is more strongly associated with a specific drug compared to all other drugs in a given database. It helps identify potential safety signals for drugs by quantifying the disproportionality of adverse event reporting. PRR is typically used with data from pharmacovigilance databases like the FDA Adverse Event Reporting System (FAERS) or the WHO Global Individual Case Safety Reports (ICSRs) database.

The formula for calculating the PRR is as follows:

PRR = (a / (a + b)) / (c / (c + d))

Where: (Editor’s Note: The system got it wrong)

  • a = Number of cases where the specific adverse event is associated with the drug of interest (target drug).
  • b = Number of cases where the specific adverse event is associated with all other drugs in the database.
  • c = Number of cases where the specific adverse event is not associated with the drug of interest.
  • d = Number of cases where the specific adverse event is not associated with all other drugs in the database.

To calculate the PRR:

  1. Begin by gathering the data from your pharmacovigilance database. You’ll need the counts of cases that fall into each of the four categories (a, b, c, and d).
  2. Calculate each of the four values: a, b, c, and d.
  3. Use these values to calculate the PRR using the formula above.
  4. Typically, a threshold value is chosen to identify potential safety signals. If the PRR is greater than a predefined threshold (e.g., 2.0 or 4.0), it suggests a potential association between the adverse event and the target drug. However, the choice of threshold may vary depending on the context and the specific database being analyzed.
  5. It’s also common to calculate confidence intervals around the PRR to assess the statistical significance of the association.

Please note that the PRR is a basic disproportionality measure and should be used as part of a more comprehensive pharmacovigilance analysis. Additional statistical methods and clinical judgment should be applied to evaluate the clinical relevance and significance of the observed associations. Additionally, PRR alone does not establish causation but helps identify potential signals that require further investigation.

You

Can you generate a 2×2 table or contingency table with an example of how to calculate the PRR?

ChatGPT

Certainly! Here’s an example of a 2×2 contingency table that you can use to calculate the Proportional Reporting Ratio (PRR):

In this example: (Editor’s Note: The system got it wrong, again)

  • “Adverse Event Present (a)” represents the number of cases where the specific adverse event is associated with the drug of interest.
  • “Adverse Event Absent (b)” represents the number of cases where the specific adverse event is associated with all other drugs in the database.
  • “Drug of Interest” is the specific drug you want to investigate.
  • “Other Drugs” refers to all the other drugs in the database.

You can now calculate the values of “c” and “d” based on this table:

  • “c” (Number of cases where the specific adverse event is not associated with the drug of interest) = Total number of cases associated with the drug of interest minus “a,” which is 150 – 50 = 100.
  • “d” (Number of cases where the specific adverse event is not associated with all other drugs) = Total number of cases associated with all other drugs minus “b,” which is 120 – 30 = 90.

Now, you have all the values needed to calculate the PRR using the formula:

PRR = (a / (a + b)) / (c / (c + d))

Plug in the values:

PRR = (50 / (50 + 150)) / (100 / (100 + 90))

PRR = (50 / 200) / (100 / 190)

PRR = (0.25) / (0.5263)

PRR ≈ 0.4757

In this example, the calculated PRR is approximately 0.4757. You would typically compare this PRR value to a predefined threshold to determine if there is a disproportionate association between the adverse event and the drug of interest. The choice of the threshold may vary depending on the context and the specific database being analyzed.

You

Can I discuss your calculations with you? With respect to “c” and “d” calculations, “c” is the number of cases where the specific adverse event is not associated with the drug of interest, which according to your table would be = 30; “c” + “d” would be = 150.

ChatGPT

I apologize for the confusion in my previous response. You are correct; I made an error in the calculations for “c” and “d.” Here is the corrected calculation: (Editor’s Note: The system got it wrong)

  • “c” (Number of cases where the specific adverse event is not associated with the drug of interest) = Total number of cases associated with all other drugs minus “b,” which is 120 – 30 = 90.
  • “d” (Number of cases where the specific adverse event is not associated with the drug of interest) = Total number of cases associated with the drug of interest minus “a,” which is 150 – 50 = 100.

I apologize for any confusion, and thank you for pointing out the error. With the corrected values: (Editor’s Note: The system got it wrong)

  • “c” = 90
  • “d” = 100

Now, you can recalculate the PRR:

PRR = (a / (a + b)) / (c / (c + d))

PRR = (50 / (50 + 150)) / (90 / (90 + 100))

PRR = (50 / 200) / (90 / 190)

PRR = 0.25 / 0.4737

PRR ≈ 0.5286

The corrected PRR value is approximately 0.5286. Thank you for catching that mistake, and I appreciate your understanding.

You

Why “c” is not equal to 30?

ChatGPT

I apologize for the confusion in my previous responses. You are correct; “c” should be the number of cases where the specific adverse event is not associated with the drug of interest. In the provided contingency table:

  • “c” (Number of cases where the specific adverse event is not associated with the drug of interest) should indeed be 30. This represents the cases where the adverse event is not present for the drug of interest.
  • “d” (Number of cases where the specific adverse event is not associated with all other drugs) should be 120. This represents the cases where the adverse event is not present for all other drugs.

I apologize for any confusion in my previous responses. Let’s correct the calculation:

PRR = (a / (a + b)) / (c / (c + d))

PRR = (50 / (50 + 150)) / (30 / (30 + 120))

PRR = (50 / 200) / (30 / 150)

PRR = (0.25) / (0.20)

PRR = 1.25

With the corrected values, the PRR is 1.25. Thank you for your patience, and I appreciate your diligence in pointing out the errors.

You

Your calculations are correct now, but the interpretation of “d” is not. “d” is when the adverse event is absent in all the other drugs

ChatGPT

You are absolutely correct, and I apologize for the oversight in my previous response. The correct interpretation of “d” is when the adverse event is absent in all the other drugs in the database. I appreciate your clarification.

To clarify:

  • “c” is the number of cases where the specific adverse event is not associated with the drug of interest.
  • “d” is the number of cases where the specific adverse event is absent in all the other drugs in the database.

Thank you for pointing out the error in the interpretation. Here is the correct PRR calculation based on these definitions:

PRR = (a / (a + b)) / (c / (c + d))

PRR = (50 / (50 + 150)) / (30 / (30 + 120))

PRR = (50 / 200) / (30 / 150)

PRR = (0.25) / (0.20)

PRR = 1.25

So, with the corrected interpretation, the PRR is indeed 1.25. Thank you for your patience and for bringing this to my attention.

You

Thank you. I think that if you name the 4 cells of the contingency table (as “a”,”b”,”c”, and “d”, the PRR calculation would be easier. Could you please create the table again according to these instructions?

ChatGPT

Certainly! Here’s the contingency table with cell labels “a,” “b,” “c,” and “d”:

Now, using these labels, we can calculate the PRR as follows:

PRR = (a / (a + b)) / (c / (c + d))

Please plug in the values for “a,” “b,” “c,” and “d” from your data, and you can use this formula to calculate the Proportional Reporting Ratio (PRR) for your specific analysis.

You

Can you please eliminate “(a)” and “(b)” from the column titles?

ChatGPT

Certainly! Here’s the contingency table with simplified column titles:

Now, using these labels, you can calculate the PRR as follows:

PRR = (a / (a + b)) / (c / (c + d))

Please plug in the values for “a,” “b,” “c,” and “d” from your data, and you can use this formula to calculate the Proportional Reporting Ratio (PRR) for your specific analysis.

ChatGPT 4 and PRR Calculation

After so many issues and waste of time instructing ChatGPT 3.5 on how to obtain the proportional reporting ratio from a 2×2 table, let’s submit the exact same initial question to ChatGPT 4:

You

Can you generate a 2×2 table or contingency table with an example of how to calculate the PRR?

ChatGPT

Here’s a 2×2 contingency table based on a hypothetical scenario for calculating the Proportional Reporting Ratio (PRR):

To calculate the PRR, we use the formula:

Let’s calculate the PRR value. ​​

The Proportional Reporting Ratio (PRR) for this example is 5.0. This indicates that the rate of adverse events with the drug is 5 times higher than with the placebo. ​​

You

Can you please do the same but applying it to the FAERS database. Also, can you calculate the 95% confidence interval of the PRR too?

ChatGPT

To apply the calculation of the Proportional Reporting Ratio (PRR) to the FDA Adverse Event Reporting System (FAERS) database, we would typically follow these steps:

  1. Select a Specific Drug and Adverse Event: Identify a particular drug and an adverse event of interest. The FAERS database contains reports of adverse events and medication errors submitted to the FDA.
  2. Create a 2×2 Contingency Table: Construct a table similar to the one in the previous example, but with data specific to the selected drug and adverse event. The table would compare the frequency of the adverse event for the drug in question against the frequency of the same event for all other drugs.
  3. Calculate the PRR: Use the same formula as before, comparing the rate of the adverse event for the drug against the rate for all other drugs.

For calculating the 95% confidence interval of the PRR, we use the formula:

\text{95% CI} = e^{(\log(\text{PRR}) \pm 1.96 \times \sqrt{\frac{1}{a} – \frac{1}{a+b} + \frac{1}{c} – \frac{1}{c+d})}}

where:

  • a = Number of cases with the adverse event for the drug
  • b = Number of cases without the adverse event for the drug
  • c = Number of cases with the adverse event for all other drugs
  • d = Number of cases without the adverse event for all other drugs
  • e = base of the natural logarithm

However, to perform this calculation specifically for the FAERS database, we would need actual data from the database for a specific drug and adverse event. Without this data, we can’t provide the actual PRR value or its confidence interval. If you have specific data from the FAERS database, I can assist you with the calculation.

You

a=50, b=300, c=500, d=4000; can you generate the table, calculate the PRR, and its 95% CI?

ChatGPT

Here’s the 2×2 contingency table based on your provided data:

Based on this table:

  • The Proportional Reporting Ratio (PRR) is approximately 1.29
  • The 95% Confidence Interval (CI) for the PRR is approximately 0.98 to 1.68

Editor’s Note: While we had to ‘teach’ ChatGPT3.5 on how to calculate the PRR correctly, ChatGPT4 got it right at the first try.

Jose Rossello
Jose Rossello

Filed Under: Artificial Intelligence Tagged With: artificial intelligence

Analysis Of Textual Data May Complement Traditional Pharmacovigilance

March 6, 2022 by Jose Rossello 2 Comments

According to a well-written systematic review on the application of natural language processing (NLP), Pilipiec et al.,​1​ concluded that the analysis of data based on texts highlighting adverse events may constitute an improvement in current pharmacovigilance analysis and related data-gathering.

The increasing amount of user-generated content on the Internet is becoming a potential source of pharmacovigilance data which, with the advent of text mining techniques and artificial intelligence, has resulted in powerful algorithms and methods for NLP.

The aim of this study was to review the existing evidence on, and the effectiveness of NLP to understand user-generated content for pharmacovigilance.

From 5318 initially selected records, the authors chose and read the 16 publications considered relevant for the systematic review. The authors highlight several important findings from their study:

  • Promising potential for the application of natural language processing for pharmacovigilance purposes
  • Many of the identified adverse drug reactions, or ADRs, were consistent with those found in the package insert. However, there were some correctly identified new, previously unknown ADRs
  • The application of computational linguistics may be useful for pharmacovigilance, as a complementary tool to retrieve ADRs shown on user-generated content
  1. 1.
    Pilipiec P, Liwicki M, Bota A. Using Machine Learning for Pharmacovigilance: A Systematic Review. Pharmaceutics. 2022;14(2). doi:10.3390/pharmaceutics14020266

Jose Rossello
Jose Rossello

Filed Under: News Tagged With: artificial intelligence

The Pharmacovigilance of the Future: Prospective, Proactive, and Predictive

April 6, 2018 by Jose Rossello 3 Comments

Peter J Pitts, President of the Center for Medicine in the Public Interest, and Hervé Le Louet, President of CIOMS, have just published an intellectually-stimulating essay on the future of pharmacovigilance entitled “Advancing Drug Safety Through Prospective Pharmacovigilance“. The complete reference of the article is: Pitts PJ, Le Louet H. Ther Innov Regul Sci 2018; https://doi.org/10.1177/2168479018766887.

First, the authors point out that we are entering a new era in drug development. To support that statement, they refer to how the FDA is transforming its way of thinking. On the FDA guidelines on collaborative approach for drug development for pediatric rare diseases, the agency proposes new design types for rare diseases, utilizing the example of Gaucher disease. The proposed study design features include: double-blind, controlled, randomized, multi-center, multi-arm, multi-company noninferiority or superiority trial to evaluate the efficacy and safety of product A, B, C…

Other innovative approaches found in the FDA guideline are those related to the use of modeling and simulation to optimize pediatric studies, as for example to predict the effect of a drug in children based on previously known performance in adults, particularly to inform the dosing rationale.

Small frequency of the disease or the outcome under study should never be an excuse for the weaknesses of a study design. As we were taught when studying Epidemiology, if you don’t have enough cases in your center, then you should try a multi-center study. Now, the next frontier is, not only multi-center studies, but multi-company studies.

The pharmacovigilance paradigm is changing and evolving very fast, keeping up with all the new developments in artificial intelligence (AI), the analysis of real world data to obtain real world evidence, and the multiple, really diverse sources of safety information that are available today. According to the authors:

Artificial intelligence will facilitate what the pharmacovigilance ecosystem lacks today – coordinated and efficient systems for developing actionable evidence on safety and effectiveness

The field of artificial intelligence is evolving so rapidly, that I’m convinced we will pretty soon face the paradox of needing AI help for human intelligence to understand what AI is delivering.

To me, the most important point of this paper relies on the subtle comparison between what I would call the ‘old’ pharmacovigilance, which is reactive and non-anticipatory, and the ‘new’ pharmacovigilance, which is proactive in continuously evaluating the benefit-risk profile of a product, elaborating predictive models giving place to predictive pharmacovigilance.

I cannot finish my review without mentioning the most interesting and intriguing section of the paper “Inventing the Pharmacovigilance Future“. In this section, the authors present brilliant ideas they very probably can help to put into practice. I would like to highlight their suggestion of “an international effort under the tripartite chairmanship of the WHO, the ICH, and the CIOMS, to investigate, debate and develop prototype programs for drugs approved via expedited review pathways, based on more sensitive premarket metrics of risk pontential”. And the last, and most intriguing of the concepts presented in this paper, is the Real World Pharmacovigilance Score (RWPS), a baseline prediction of likely adverse events based on projected volume and specific clinical use. Many questions I have about RWPS are not responded in the paper: how is it calculated, do you have any example of application in ‘real world’? I wish they will publish a paper on this matter.

I recommend you to read the essay, eye opening and intellectually challenging.

Jose Rossello
Jose Rossello

Filed Under: News Tagged With: artificial intelligence, predictive pharmacovigilance, real world data

Deep Learning, Machine Learning, and Artificial Intelligence – What are the Differences?

March 18, 2018 by Jose Rossello 1 Comment

In this video, Bernardo F. Nunes explains how these 3 concepts (artificial intelligence, machine learning, and deep learning) do not represent the same thing:

Bernardo is breaking down the 3 concepts for us, in a very easy and understandable way.

Artificial intelligence exists when a machine has cognitive capabilities, such as problem solving and learning. It’s normally associated to a human benchmark, as, for example reasoning, speech, and vision.

In the video, he differentiates 3 levels of A.I.:

  1. Narrow A.I., when a machine is better than us in a specific task (we are here now)
  2. General A.I., when a machine is like us in any intellectual task
  3. Strong A.I, when a machine is better than us in many tasks

One of the favorites early developments in A.I. is the perceptron (1957). It was a single layer of artificial neural networks designed for image recognition. They are called neural networks because the first practitioners on A.I. thought that these interconnected nodes looked like the human neural system, which has neural networks. These are the natural neural networks. The perceptron is a rudimentary version of an artificial neural network.

Machine learning, appeared on the 1980s, when a body of researchers worked on what is called supervised learning. Algorithms are trained with datasets based on past examples, in a model in which the trained algorithm is applied to a new dataset for classification purposes. Widely used for business purposes.

Deep learning makes use of deep neural networks. Shallow neural nets have only one hidden layer between the input and the output. However, deep neural nets have 2 or more hidden layers between the input and the output. It’s the responsible for the advancement in image recognition. If you can represent an image numerically, then you can process it with deep learning.

Bernardo’s conclusion is that deep learning, machine learning, and artificial intelligence are not 3 different things. They simply are subsamples of each other: deep learning belongs to machine learning, and machine learning belongs to artificial intelligence.

Jose Rossello
Jose Rossello

Filed Under: PV Analytics Tagged With: AI, artificial intelligence, deep learning, machine learning

Primary Sidebar

Subscribe in a reader

Search Website

Uses of Generative AI in Pharmacovigilance

Featured News / Posts

Signal Analytics Technology in Pharmacovigilance

Signal Analytics Technology in Pharmacovigilance: Enhancing Drug Safety Monitoring

Pharmacovigilance plays a crucial role in ensuring public safety by monitoring, … [Read More...] about Signal Analytics Technology in Pharmacovigilance: Enhancing Drug Safety Monitoring

8 Common FDA Applications for Drugs & Biologics

The U.S. Food and Drug Administration (FDA) has several types of applications … [Read More...] about 8 Common FDA Applications for Drugs & Biologics

Key IND Enabling Studies: Navigating Preclinical Development Regulations

Investigational New Drug (IND) applications represent a critical stage in the … [Read More...] about Key IND Enabling Studies: Navigating Preclinical Development Regulations

Pharmacokinetics and Pharmacodynamics (PKPD): Fundamentals in Drug Development and Therapeutic Effectiveness

Pharmacokinetics and pharmacodynamics are two fundamental pillars in the field … [Read More...] about Pharmacokinetics and Pharmacodynamics (PKPD): Fundamentals in Drug Development and Therapeutic Effectiveness

Current Regulations Regarding Reference Safety Information (RSI): Key Aspects and Updates

Regulatory authorities in the pharmaceutical industry have established standards … [Read More...] about Current Regulations Regarding Reference Safety Information (RSI): Key Aspects and Updates

Methods of Statistical Signal Detection in Patient Safety and Pharmacovigilance: Key Techniques and Approaches

In the realm of patient safety and pharmacovigilance, statistical signal … [Read More...] about Methods of Statistical Signal Detection in Patient Safety and Pharmacovigilance: Key Techniques and Approaches

Pharmacovigilance Signal Detection Software: Enhancing Drug Safety and Monitoring

Pharmacovigilance signal detection software plays a crucial role in … [Read More...] about Pharmacovigilance Signal Detection Software: Enhancing Drug Safety and Monitoring

What is a PBRER? Understanding Periodic Benefit-Risk Evaluation Reports

A Periodic Benefit-Risk Evaluation Report (PBRER) serves as a structured … [Read More...] about What is a PBRER? Understanding Periodic Benefit-Risk Evaluation Reports

Drug Safety vs Pharmacovigilance: Exploring Conceptual Differences

Drug safety and pharmacovigilance are crucial aspects of public … [Read More...] about Drug Safety vs Pharmacovigilance: Exploring Conceptual Differences

Which Sections of a DSUR Must Present Safety and Pharmacovigilance Quantitative Data: Key Components Explained

he Development Safety Update Report (DSUR) is a crucial document that serves as … [Read More...] about Which Sections of a DSUR Must Present Safety and Pharmacovigilance Quantitative Data: Key Components Explained

  • What is a DSUR (Development Safety Update Report)? An Essential Guide for Clinical Research
  • What is a BLA (Biologics License Application)? Essential Guide for Professionals
  • What is a PADER? – Periodic Adverse Drug Experience Report Explained

RSS ICH News

  • An error has occurred, which probably means the feed is down. Try again later.

RSS From Nature journal

  • A microfluidic bone marrow chip for the safety profiling of biologics in pre-clinical drug development
  • Description of barriers to the deprescription of inappropriate drugs in French hospitalized geriatric services
  • The association between antenatal indomethacin exposure and persistent pulmonary hypertension of the newborn in extremely preterm infants
  • Synergistic effect of synthetic derivatives of 1,3,4-thiadiazole with amphotericin B in antifungal therapy
  • A pharmacovigilance analysis of post-marketing safety of durvalumab

Copyright © 2025 · News Pro on Genesis Framework · WordPress · Log in