Skip to main content
search

October 6, 2025

How can the latest modeling and simulation technologies accelerate therapeutic agent discovery and development?

Model-informed drug development (MIDD) is critical to informing the development of safer and more effective new drugs. MIDD can be used to assess drug performance, support regulatory and payor success, characterize diseases, and optimize clinical trial designs.

This blog will focus on best practices for model-informed drug development in oncology.

MIDD maximizes and connects the data collected during non-clinical and clinical development and real-world settings, consisting of individual-level and summary-level information. Learn more about our real-world evidence services here.

It also enables extrapolation to unstudied situations and populations, helping to anticipate the potential risks, mitigate them, and improve the probability of success. Using MIDD can accelerate timelines by bridging across development phases to increase development certainty and leverage existing knowledge and improve cost-effectiveness.

Why is MIDD so powerful?

Maximizes the information from gathered data

Confidence in drug
Confidence in target
Confidence in endpoints
Confidence in regulatory decisions
Drug performance assessment, disease characterization, clinical trial optimization, regulatory & payor success

Allows extrapolation to new situations

  • Support for safety & efficacy of doses not studied
  • Predictions of behavior in special populations
    • Elderly & children
    • Renal & hepatic impairment
  • Avoid unnecessary studies
    • DDI studies
    • BE studies
  • Combination selections

Top-down (MIDD approaches)

MIDD has become an established approach in the last decade. However, pharmacokinetic/pharmacodynamic (PK/PD) and population PK/PD (PopPK/PD) modeling and simulation (M&S)/pharmacometrics techniques were introduced over 60 years ago. In the 90s, they were largely used experimentally to support drug development programs with limited impact on decision-making. From 2000 to 2010, using PopPK and PK/PD became embedded in drug development and is now critical to many international regulatory guidance documents and frameworks. In addition, global regulatory agencies also encourage integrating MIDD approaches into drug submissions, with increasing recognition of Model Informed Precision Dosing (MIPD) to support precision dosing strategies 

Understanding how an investigational drug’s safety and efficacy profile compares to the standard of care and/or competitor drugs is important medically and commercially. 

Model-based meta-analysis (MBMA) uses highly curated clinical trial data in databases (e.g. Codex) and literature and combined with pharmacometrics models to enable indirect head-to-head comparison, considering the impact of treatment, dosing regimen, patient population, and trial characteristics on responses to medicines. This can support designing and executing pivotal studies. In some cases, MBMA models can even serve as an external control arm. 

Bottom-up (mechanistic MIDD approaches)

Large pharma adopted mechanistic modeling approaches such as physiologically-based pharmacokinetic (PBPK) modeling and simulation, quantitative systems pharmacology (QSP), and semi-mechanistic PK/PD modeling as experimental techniques in the 90s. Nowadays, PBPK modeling to predict first-time-in-human-dosing, drug-drug interactions (DDIs) and explore dosing in special populations, such as pediatric, pregnant, and lactating populations, has become the pharmaceutical industry standard. These models are also frequently used to predict PK in unstudied populations. 

In recent years, the use of QSP has increased. QSP combines computational modeling and experimental data to examine the relationships between a drug, the biological system, and the underlying disease process. By combining mechanistic understanding of molecular pathways with pharmacokinetic and pharmacodynamic data, QSP enables researchers to predict drug effects across different patient populations, explore novel combination therapies, and identify biomarkers for response or toxicity. This approach supports decision-making throughout drug development, from target identification and lead optimization to clinical trial design and dose selection. 

Finding the right tool for the job

QSP
  • New modalities
  • Dose selection & optimization
  • Combination therapy
  • Target selection
  • Safety risk qualification
PK/PD
  • Dose-response relationship
  • Drug exposure
  • Subject variability
  • Dose regimen
QSP, PK/PD, PBPK, MBMA
PBPK
  • Drug-drug interactions
  • Special populations
  • Formulation development
  • FIH dosing
  • In silico dermal bioequivalence
MBMA
  • Comparator analysis
  • Trial design optimization
  • Bridging
  • Go/no decisions

From “nice to have” to “regulatory essentials”

In recent decades, PopPK, PK/PD, and exposure-response MIDD have become expected components for late-stage clinical drug development programs. These “pharmacometric” approaches are also accepted as standard regulatory development tools and included in development planning. These activities are no longer considered a nicety. They’re a necessity/pre-requisite! Global regulatory agencies expect drug developers to apply these tools throughout a product’s life cycle where it’s feasible to support key questions for decision-making and validating assumptions to minimize risk. 

MIDD isn’t just used to improve decision-making. It is also increasingly used to improve the likelihood of achieving desired outcomes while maximizing safety and efficacy of development programs, to optimize clinical study designs, and even to minimize the scale. Ultimately, using MIDD may even support waivers for conducting certain clinical studies. 

Model-informed drug development in oncology drug development

MIDD can be applied to all therapeutic areas and various modalities. This blog highlights its application to oncology therapeutics development. This approach can inform all stages of cancer drug development from early to late clinical development. It allows us to understand an investigational oncology drug’s PK, safety, and efficacy, first in nonclinical species. Then this information can be translated to first-in-human studies where it can inform the dosing strategy (dose, dosing intervals, dose schedule, and dose escalation/de-escalation plan). In addition, PK/PD modeling can help characterize the impact of intrinsic (age, weight, sex, genetic factors, mutations etc.) and extrinsic (food, co-medications, smoking status, laboratory variables etc.) covariates on drug exposure. Concentration-ECG (e.g., C-QTc) analysis using early-stage clinical trial PK-matched QTc data can de-risk the cardiac safety risk or waive the need for a clinical TQT study.  

Oncology drugs are often combined with novel compounds or the standard of care. However, choosing the optimal drug combinations is difficult using conventional clinical trial designs. Emerging mechanistic approaches, such as mechanistic compartmental and QSP modeling, have enabled us to assess downstream biochemical and cellular effects of modulating the target pathway to support developing a drug combination and safety strategy. Lastly, MBMA can be used to help support the optimal trial design, to understand the competitive landscape and in some cases to create an in silico external control arm.  

Ultimately, using MIDD approaches can save time and money while increasing drug efficacy and minimizing risk to the patients.

References

Jansson-Löfmark R, Fridén M, Badolo L, et al. Translational PK/PD: a retrospective analysis of performance and impact from a drug portfolio. Drug Discov Today. 2025;30(7):104417. doi:10.1016/j.drudis.2025.104417 

Sahasrabudhe V, Nicholas T, Nucci G, Musante CJ, Corrigan B. Impact of Model-Informed Drug Development on Drug Development Cycle Times and Clinical Trial Cost. Clin Pharmacol Ther. 2025;118(2):378-385. doi:10.1002/cpt.3636 

FAQs

What does Pharmacokinetics / Pharmacodynamics (PK/PD) refer to?

PK describes what the body does to a drug (absorption, distribution, metabolism, excretion), whilst PD describes what the drug does to the body (the drug’s effect). PK/PD models link drug concentration to its effect.

What is Population Pharmacokinetics (PopPK) modeling and simulation?

This is a type of modeling that analyzes sources of variability in drug concentrations within and between individuals in a patient population to understand how factors like age, weight, or genetics affect a drug’s pharmacokinetics. The statistical approach uses sparse sampling (i.e. a few blood samples per patient, taken on more than one occasion) which allows the technique to be applied to therapeutic studies.

What is physiologically-based pharmacokinetic (PBPK) modeling?

This is a mechanistic modeling approach that simulates how a drug moves through and is processed by different organs and tissues in the body based on physiological, biochemical, and drug-specific properties. It’s often used to predict drug-drug interactions (DDIs) and outcomes in unstudied populations (e.g., pediatrics, pregnant women, patients with renal/hepatic impairment).

How can MIDD support the FDA's roadmap to eliminate animal testing?

MIDD is key to the shift away from animal testing, offering robust alternatives that align with the FDA’s roadmap and the “3Rs” principle (reduce, replace, refine). For example, techniques like PBPK and QSP modeling use physiological and biochemical data to predict drug behavior, drug-drug interactions, and dosing in humans—without animal testing. By creating virtual models of patients and diseases, MIDD reduces reliance on animal testing while improving the accuracy of preclinical translation. This paves the way for drug development that is faster, cheaper, and more humane.

What is the business impact of MIDD on the pharmaceutical industry?

MIDD has a significant impact on the pharmaceutical industry by streamlining drug development, cutting costs, and speeding up timelines. By combining data from non-clinical and clinical studies, MIDD enables better decision-making and greater certainty across all development phases. For example, a study conducted by Pfizer found that systematic use of MIDD saves an average of 10 months per program. In another study, AstraZeneca found that mechanism-based biosimulation increased the chances of achieving a positive proof of mechanism by 2.5 times, helping to reduce risks and focus resources on the best candidates. Watch this video about how MIDD increases the efficiency of pharma R&D  to learn more. 

Dr. Fran Brown

Senior Vice President, Drug Development Science

Dr. Fran Brown is a highly respected professional with proven leadership skills and 28 years of broad experience within pharmaceutical development and due diligence. She has extensive experience with strategic and operational global drug development from early discovery to filing and post-marketing. This experience spans multiple therapeutic areas, small molecules and biologics, global regulatory requirements and registration pathways. She possesses a broad knowledge of product development and portfolio management, with a special focus on development strategy, regulatory interactions and product filings.

Her past appointments include leadership roles within large Pharma as well as in small biotech organizations including head of clinical pharmacology, clinical leader, project development leader, head of clinical operations and due diligence asset assessment. She has over 10 years of experience in providing consulting advice to the pharmaceutical industry and non-profit Global Health Organizations ranging from individual project support, to strategic TA strategy and development planning, portfolio management and corporate transformation. She joined Certara in 2017 and is currently the SVP of Drug Development Science within Integrated Drug Development.

Amy Cheung, PhD, Vice President, Certara Drug Development Solutions at Certara
Dr. S.Y. Amy Cheung

Vice President, Quantitative Science Service

S. Y. Amy Cheung is Vice President, Certara Drug Development Solutions. Dr. Cheung has over a decade of experience working in the pharmaceutical industry at AstraZeneca (AZ), with her role as Senior Pharmacometrician and Project manager of AZ Paediatric working group. She obtained her Ph.D. from the University of Manchester, on the topic of Structural Identifiability Analysis in Pharmacokinetic and Pharmacodynamic Models. After receiving her Ph.D. she worked as a postdoc on mechanistic modeling at the Centre for Applied Pharmacokinetic Research (CAPKR) at the University of Manchester.

She was the co-lead for the cardiac safety training for the IMI DDmoRe project and is also an active member of the EFPIA Model Informed Drug Discovery and Development (MID3) workgroup. She was a chair of IQ Consortium Clinical Pharmacology Leadership Group Pediatric Working Group in 2018 and current co-chair of IQ Consortium TALG, CPLQ PBPK Pediatric group.

This blog was originally published in September 2021 and has been updated for accuracy and comprehensiveness.

Our experts have decades of experience enhancing and optimizing the drug development process

At Certara, we consult with you to address all your drug development challenges. We can streamline every stage of development by providing a tailored solution that combines model informed drug development with expertise across due diligence, CMC, toxicology, DMPK, regulatory strategy, clinical pharmacology, modeling & simulation, biometrics, and more. By uniting data, insights, and expert-driven strategies, we help you deliver new therapies faster to patients.

Learn more about our drug development services

Contact Certara