Ca-ssis/istock via Getty Images
Role of advanced modeling and virtual data in drug approvals
Advanced modeling and virtual data help biopharma companies design efficient, ethical and cost-effective trials to accelerate drug development and regulatory approvals.
At the American Association of Pharmaceutical Scientists 2024 PharmSci 360 conference in Salt Lake City, Vibha Jawa, executive director of bioanalytics at Bristol Myers Squibb, presented an innovative perspective on the role of advanced modeling and virtual data in clinical trials. Her prologue presentation highlighted how combining empirical and mechanistic models, virtual data and synthetic controls reshapes drug development processes, accelerates timelines, and refines regulatory approaches.
Using these tools, biopharma companies can achieve more efficient, ethical and impactful trials, accelerating the safe delivery of innovative therapeutics to market.
Empirical and mechanistic models in drug development
In traditional pharmaceutical development, empirical models have long provided foundational support, helping researchers predict basic pharmacokinetics and pharmacodynamics without delving into mechanistic principles.
"Empirical models are mathematical formulas based on observed data without underlying mechanistic principles," Jawa described.
The simplicity of these models makes them particularly useful in early-stage drug development. Researchers can focus on essential aspects like drug absorption, concentration and bioavailability based on collected data.
However, empirical models have inherent limitations. Since they rely on observed data, they often cannot predict outcomes beyond the data set they are built on, restricting their utility in more complex scenarios.
"Empirical models are sensitive to assumptions and lack the ability to simulate a drug's mechanistic effects, such as its interactions with physiological matrices," Jawa explained.
This gap has led researchers to develop and employ mechanistic models, which simulate drug action by incorporating biological, physiological and pharmacological factors. These models provide a richer understanding of drugs' behavior within biological systems.
Unlike empirical models, mechanistic models allow researchers to explore drug disposition and optimize dosage regimens to increase therapeutic impact.
"Mechanistic models allow us to simulate the behavior of a drug, optimize therapeutic outcomes and understand the mechanism of action," Jawa noted.
She explained that by blending empirical data with mechanistic insights, researchers gain a more comprehensive toolkit that bolsters predictive power, aiding in developing personalized medicine approaches.
Integrating empirical and mechanistic models
A major strength of mechanistic models lies in their predictive capabilities, allowing researchers to simulate how drugs might behave across diverse patient populations and in different disease states. Jawa outlined how physiologically based pharmacokinetic (PBPK) models can simulate drug absorption, metabolism, and excretion while incorporating physiological parameters like blood flow rates, organ size, patient demographics and disease severity.
Vibha JawaExecutive director of bioanalytics, Bristol Myers Squibb
"PBPK models enable us to account for demographic differences, drug-drug interactions and physiological variations, making them a powerful tool for dose optimization and personalized medicine," Jawa emphasized.
This integration of empirical and mechanistic models also allows for a more holistic assessment of potential drug effects, a particularly valuable feature in complex conditions. Quantitative systems pharmacology models, for example, integrate cellular, molecular, and tissue-level mechanisms, providing a multilayered view of drug responses.
"These models capture complex interactions and feedback loops within biological systems, bridging preclinical and clinical stages," Jawa mentioned, highlighting their potential for developing disease progression models and personalized medicine strategies.
With empirical models providing a simple, assumption-limited framework and mechanistic models enabling in-depth simulations, their combined use creates a powerful system that overcomes the limitations of each model when used alone. Jawa noted that case studies demonstrate how these complementary approaches help identify optimal dosing regimens, anticipate potential adverse effects and adapt dosing to individual patients' needs -- all of which support a new era in drug development.
Enhancing trial design and efficiency
Virtual data has emerged as a transformative element in clinical trial design, offering new ways to conduct trials where obtaining real-world data might be challenging. Jawa described virtual data as "model-generated data that mimics real-world characteristics," which can supplement limited datasets, especially in trials involving rare diseases, pediatric populations, or conditions with small patient pools.
"Virtual data allows researchers to supplement limited real data, especially in rare disease or pediatric populations," Jawa added, enabling predictive analytics and optimized trial design.
The use of virtual data extends to multiple trial phases, helping researchers perform scenario testing, identify optimal patient populations, and even test different treatment regimens and dosing schedules before committing resources to traditional clinical trials.
By conducting scenario testing in advance, researchers can better tailor study designs to maximize trial success and cost-effectiveness, a critical advantage in today's competitive biopharma environment.
"Virtual data enables optimization of trial designs and identification of the right patient populations through predictive models and analytics before implementing costly trials," Jawa emphasized.
Another key innovation is synthetic control groups, which offer viable alternatives when traditional control groups might be infeasible or unethical, such as in trials for last-line treatments. Synthetic controls combine data from historical studies or real-world data sets to emulate a control group, providing a robust comparative basis for new therapies.
According to Jawa, "Synthetic controls closely resemble characteristics of the treated group, filling gaps where traditional controls aren't feasible."
Synthetic control groups allow for accurate comparisons while addressing ethical concerns about withholding treatment from certain populations. Applications of synthetic controls extend beyond patient ethics; they also help researchers optimize resources, reduce trial costs and enhance statistical power by improving comparison precision. These groups are especially useful in rare disease and oncology trials, where traditional placebo-controlled trials might not be viable.
"Synthetic controls can supplement real-world evidence and enhance statistical power, increasing the likelihood of detecting meaningful outcomes," Jawa said, underscoring their value in complex studies where traditional controls might not be available.
RWD's growing impact on modeling and regulatory acceptance
Real-world data (RWD) has quickly become indispensable in drug development, providing a broader perspective on treatment effectiveness, safety profiles and long-term outcomes. With sources ranging from electronic health records to wearable devices, RWD complements randomized controlled trials by adding insights into real-world patient behavior and responses to treatment.
"Real-world data can support regulatory decisions by providing additional evidence on post-marketing safety and efficacy, facilitating label expansions, and regulatory solutions," Jawa highlighted.
RWD can also guide hypothesis generation and treatment optimization, helping researchers better understand disease progression and patient characteristics. Despite these benefits, RWD poses challenges due to its inherent heterogeneity across patient demographics, treatment settings and disease states. Quality assurance and validation are crucial to overcoming these challenges, as is a rigorous focus on data consistency and relevance.
"The heterogeneity in real-world data must be carefully managed to maintain data quality and generalizability," Jawa advised, emphasizing that well-validated RWD can significantly strengthen submissions for regulatory approval.
The future of modeling in drug development
With advanced modeling, virtual data and synthetic controls in place, the pharmaceutical industry is moving toward more efficient, ethical and patient-centered drug development. Computational models allow researchers to explore alternative trial designs that avoid the ethical dilemmas of placebo use in vulnerable populations and reduce reliance on animal models.
"Computational models simulate biological and pharmacological processes, providing alternatives to traditional controls and helping to make trials more ethical," Jawa noted.
The resource efficiency gained through modeling also allows biopharma companies to accelerate timelines and reduce costs, which is vital in the push to bring innovative treatments to market faster. The FDA and other regulatory bodies have responded positively to these innovations, provided there is sufficient evidence of data quality, model validation and reliability.
Jawa underscored the importance of industry-wide collaboration. "The future acceptance of these models will depend on partnerships between researchers, regulators and industry to establish best practices and validation protocols," Jawa added.
Embracing personalized medicine and adaptive trial design
The ultimate promise of these advanced methodologies lies in their potential to support personalized medicine, early access programs and adaptive trial designs. Personalized medicine, in particular, benefits from the ability of PBPK and other mechanistic models to adjust dosing regimens and therapeutic approaches to individual patient characteristics.
"This is where we are heading next," Jawa suggested, "personalized medicine, delivering treatments based on individual patient characteristics and their real-world responses."
Advanced modeling and virtual data can reshape the regulatory landscape and improve patient outcomes by reducing costs, optimizing trial designs, and generating more reliable data. With further research, industry collaboration, and regulatory endorsement, these methodologies are expected to become integral to the next generation of drug development. Jawa's prologue presentation at the American Association of Pharmaceutical Scientists 2024 PharmSci 360 conference clearly signals that the future of clinical trials will be one of increased precision, efficiency and ethical sensitivity, paving the way for a new era in pharmaceutical innovation.
Alivia Kaylor is a scientist and the senior site editor of Pharma Life Sciences.