跳转到主要内容
搜索

2025 年 10 月 24 日

Antibody–drug conjugates (ADCs) are among the most exciting advances in targeted therapy. By combining the precision of monoclonal antibodies with the potency of cytotoxic drugs, ADCs have transformed the way certain cancers are treated, with more than fifteen products now approved worldwide.

But the same features that make ADCs powerful also create unique challenges in pharmacokinetics (PK). Multiple analytes, variable drug–antibody ratios (DAR), nonlinear kinetics, and multiple elimination pathways mean that traditional PK approaches must be adapted. Developers need to balance complexity and feasibility while keeping things mechanistically meaningful.

This blog shares key lessons in applying noncompartmental analysis (NCA) and population PK (popPK) to ADCs—and highlights opportunities for smarter drug development strategies.

Lesson 1: Treat ADCs as multi-analyte systems

Unlike small molecules or antibodies alone, ADCs are made up of several interdependent components:

  • Conjugated antibody (ADC): the intact antibody with attached payloads, usually the main driver of efficacy.
  • Total antibody: includes both conjugated and unconjugated (“naked”) antibody.
  • Free payload or unconjucated payload: the small molecule released from the ADC, often responsible for off-target toxicity. May influence efficacy, if there’s extracellular release.
  • Antibody-conjugated payloads: reflects how much payload is attached to the antibody.

There are tight inter-analyte PK relationships. For example, free payload exposure depends on release from the ADC, while naked antibody arises from deconjugation. Treating ADCs as a single analyte risk missing these critical dynamics.

Lesson 2: Recommended to use molar units

One of the most common pitfalls in ADC analysis is reporting only in mass units. Because antibodies, payloads, and linkers differ greatly in molecular weight, comparisons in ng/mL can be misleading.

Switching to molar units makes it possible to:

  • Perform mole-balance checks.
  • Calculate clearance and volume accurately.
  • Normalize doses across analytes.
  • Compare constructs with different DAR values (e.g., DAR 4 vs DAR 8), since DAR is defined as a mole ratio.

Without this step, dose–exposure relationships and cross-study comparisons may be unreliable.

Lesson 3: DAR is central to PK, efficacy, and safety

DAR distribution shapes nearly every aspect of ADC behavior. ADC is a mixture of molecules with varying number of attached payloads, DAR species, each with a distinct PK profile.

  • Higher DAR species tend to clear more quickly and often show nonlinear clearance
  • Lower DAR species circulate longer, behaving more like unconjugated antibodies.
  • Balance is key: while higher DAR increases potency by delivering more payload per antibody, it can also reduce stability and increase off-target effects.

Accounting for DAR during NCA and popPK analysis can help understand the PK and ensures more accurate exposure metrics and avoids flawed interpretations.

Lesson 4: Adapt NCA outputs for ADCs

NCA remains a trusted tool for early PK assessment—it is fast, transparent, reproducible, and regulator-accepted. But for ADCs, the standard outputs need to go further.

  • Overlay plots of antibody vs ADC concentrations can confirm stability.
  • Correlation plots (e.g., payload vs ADC post-Tmax) show whether one analyte can reliably predict another.
  • Exposure ratio tables highlight how payload exposure compares to ADC exposure, offering insights into safety and stability.
  • Nonlinearity checks help identify target-mediated drug disposition (TMDD), which often presents as a change in half-life with concentration.

These tailored outputs provide a clearer, more complete picture of how an ADC behaves in vivo.

Lesson 5: Know when to move beyond NCA

NCA offers a snapshot of “what happened,” but more complex questions require models that can simulate “what if.”Population PK modeling becomes essential when:

  • Optimizing or comparing dosing regimens (e.g., every six vs every eight weeks).
  • Projecting dosing for pediatrics or special populations.
  • Separating efficacy drivers (intact ADC) from safety drivers (free payload).
  • Capturing nonlinearities like TMDD.
  • Assessing the impact of anti-drug antibodies (ADAs) on different analytes.

Recognizing these triggers early allows teams to move from descriptive analyses to predictive insights, supporting more confident development and regulatory decisions.

Opportunities ahead

Applying these lessons not only improves how ADCs are analyzed today, but also opens the door to new opportunities:

  • Closer assay–model integration: ensuring that what is measured in the lab maps directly to analytes needed in model development.
  • Faster pediatric access: using popPK models with allometric scaling and maturation functions.

Summary and next steps

ADCs hold tremendous therapeutic potential but require tailored pharmacokinetic approaches. Noncompartmental analysis (NCA) provides the foundation when adapted for multi-analyte systems, molar units, and DAR. Knowing when to advance to population PK modeling is equally important to refine dosing, balance safety and efficacy, and build regulatory confidence.

Success starts with understanding the ADC—its mechanism, analyte profiles, and DAR dynamics—to create models that are both robust and actionable. Applying these principles helps developers bring ADCs to patients faster and extend their benefits beyond oncology.

Erika Brooks

Marketing Director, Quantitative Science Services

With over 22 years of experience in hospitals, health systems, associations, life sciences, physician practices, and suppliers, Erika is an experienced marketing strategist and supports the Quantitative Science Services offering with Go-to market planning and execution.

Martin Beliveau

Vice President Consulting, Certara

Dr. Martin Beliveau has over 10 years of modeling experience in clinical pharmacology. He joined Certara Strategic Consulting in 2007. Before that, he was a pharmacokinetic scientist and Study Director at Charles River Laboratories. His entire career has been in the area of pharmacokinetic analysis, modeling and simulation for regulatory submissions and decision making. His work covers areas a wide range of indications with a particular focus on translational medicine, first-in-human predictions and biodefence.

Dr. Martin Beliveau received a PhD in Public Health from the Université de Montreal under Dr. Kannan Krishnan in physiologically-based pharmacokinetic modeling in 2004. Aside from his modeling/simulation activities, Martin enjoys reading a good Spider-Man story.

Eline van Maanen

Director Consulting, Certara

Dr. Eline van Maanen brings over 16 years of experience in pharmacometric consulting. She joined Certara in 2018. Before that, she was a PKPD consultant at LAP&P Consultants. Eline has dedicated her entire career to the field of pharmacometrics. Her expertise includes mechanistic PKPD modeling across a range of therapeutic areas, such as neurology, oncology and infectious diseases.

Eline received a PhD in Pharmacology from Leiden University in 2017, where she studied systems pharmacology of the amyloid cascade under the supervision of Prof. Meindert Danhof. Eline has a background in engineering, holding a MSc degree in Life Science and Technology from Delft University of Technology. Outside of work, Eline enjoys pilot-gig rowing.

Ready to take the next step? Connect with our experts to discuss your ADC program

Every antibody-drug conjugate presents unique PK challenges. Our pharmacometric and modeling experts can help you design the right approach — from NCA to population PK — to accelerate your development and de-risk decisions.

Schedule a consultation to start the conversation

FAQs: Applying NCA and PopPK to ADCs

Why are ADCs challenging to analyze?

ADCs are multi-analyte drugs made up of antibodies, payloads, and linkers—all behaving differently in the body. Their complexity creates nonlinear kinetics and multiple elimination pathways that standard PK methods don’t capture well.

What’s the difference between NCA and PopPK for ADCs?

NCA gives a quick, transparent look at how each analyte behaves—great for early studies. PopPK goes deeper, modeling across populations to predict dosing, safety, and efficacy under different conditions.

Why use molar units instead of mass units?

Molar units account for differences in molecular weight, making it easier to compare analytes, check mole balance, and evaluate DAR effects accurately.

What’s the importance of DAR?

DAR—how many payloads are attached to each antibody—drives clearance, potency, and safety. High DAR species clear faster and can increase toxicity; low DAR species circulate longer. Tracking DAR helps explain exposure and response differences.

When should you move beyond NCA?

When you need to predict outcomes, test dosing regimens, model pediatric exposure, or understand nonlinearities and ADA effects—PopPK becomes essential.

How can NCA be adapted for ADCs?

Include overlay plots of analytes, exposure ratios, and correlation checks between ADC and payload levels to get a clearer picture of stability and release dynamics.

What’s next for ADC PK modeling?

Better assay–model alignment, DAR-informed PopPK models, and pediatric extrapolations are opening new doors for faster, safer ADC development.

Where can I learn more?

Watch the on-demand webinar with Certara experts Martin Belliveau, Eline van Maanen, and Kang Lin from Avidity Biosciences for real-world examples. 立即观看

与专家对话


沪ICP备2022021526号

Powered by Translations.com GlobalLink Web Software