09/11/2025 | News release | Distributed by Public on 09/11/2025 06:12
Selecting the optimal first-in-human (FIH) dose is one of the most challenging steps in drug development. Traditionally, toxicologists and clinical pharmacologists have relied on two metrics to determine the maximum recommended starting dose (MRSD): NOAEL (no observed adverse effect level) and MABEL (minimal anticipated biological effect level). While these measures provided a starting point, experience and evolving science have exposed their limitations, especially in the era of molecular-targeting immunotherapies. Today, quantitative systems pharmacology (QSP) offers a more comprehensive approach, and partnering with experts like Simulations Plus can enhance both safety and efficacy in dose selection.
For years, MABEL and NOAEL have been the backbone of determining MRSD for FIH study designs. MABEL estimates the minimum dose that induces a measurable biological effect, while NOAEL identifies the highest dose with no observed adverse effects in preclinical studies. These metrics work reasonably well in the context of chemotherapy which has narrow therapeutic windows, where predictable dose-response relationships and established toxicity profiles make translating animal data to humans relatively straightforward.
However, the situation changes dramatically with targeted immunotherapies. Unlike traditional chemotherapeutics, many immunomodulatory agents interact with complex immune networks. Small dose variations can trigger significant, nonlinear responses, widening the therapeutic index (Figure 1). For instance, the same immunotherapy that drives an effective anti-tumor response might also induce severe adverse events, such as cytokine release syndrome. Relying solely on MABEL or NOAEL, which are empirical and based on animal data, may therefore fail to predict critical nuances of human responses.
There are three major areas of inadequacy that MABEL and NOAEL exhibit in targeted immunotherapy.
Overreliance on preclinical data: MABEL is often derived from in vitro potency assessments and scaled animal data, while NOAEL is established from non-human primate (NHP) studies. Yet, differences in physiology, metabolism, and antigen can limit the accuracy of these metrics when applied to humans, resulting in MABEL underestimating (see Figure 2) and NOAEL overestimating the MRSD. A notable example of the latter was the 2006 incident with the CD28 super-agonist antibody TGN1412, where six human volunteers became critically ill-a failure not predicted by animal models because of limited cross-species antibody reactivity. This event underscored the need for alternative methodologies such as QSP.
Static endpoints in a dynamic system: Both MABEL and NOAEL reduce complex pharmacological behavior to a single value-one indicating a minimum effective concentration and the other a toxicity threshold. They do not capture the evolving interplay between a drug's pharmacokinetics (PK) and pharmacodynamics (PD) over time, which is essential for understanding drug behavior in the human body. In immunotherapy, factors like receptor saturation, feedback mechanisms, and cross-talk among cellular pathways require a dynamic approach to dosing.
Lack of mechanistic insight: By ignoring the underlying biological networks, MABEL and NOAEL provide limited information about the drug's mechanistic action. This oversimplification can lead to underestimating off-target effects or delayed toxicities and potentially enforces a one-size-fits-all dosing strategy that might not account for individual patient variability.
QSP represents a paradigm shift in FIH dose selection. By harnessing mathematical models and computational simulations, QSP integrates PK, PD, and detailed biological mechanisms to offer a dynamic and realistic picture of drug behavior. QSP provides a better estimate of:
Chemotherapy typically follows a predictable dose-toxicity curve, where maximum tolerated doses and off-target effects are well characterized. In contrast, immunotherapies modulate complex immune responses that are less predictable. Consequently, what might be an effective dose for one patient may lead to toxicity in another. This variability highlights the limitations of one-dimensional approaches like MABEL and NOAEL, and underscores the need for sophisticated, dynamic modeling via QSP.
For companies seeking to adopt QSP, partnering with expert scientists offers significant benefits:
While MABEL and NOAEL have served as traditional tools for FIH dose determination, their limitations are increasingly evident in the context of immunotherapy. The dynamic and multifactorial nature of immune responses requires a modeling approach that goes beyond a simple threshold value. QSP, with its integration of mechanistic insights and dynamic simulations, not only bridges the translational gap but also offers the flexibility needed for personalized therapy.
For drug developers, embracing QSP means moving away from the one-size-fits-all mindset, toward a more nuanced understanding of dosing that can enhance both patient safety and therapeutic efficacy. As immunotherapies continue to evolve, partnering with QSP experts becomes essential for unlocking the full potential of innovative treatments. This shift towards QSP promises to pave the way for next-generation clinical trials-safer, faster, and more attuned to the complexities of human biology.
By integrating advanced simulation tools and embracing model-informed strategies, the future of FIH dosing is not only more predictable but also tailored to the individual needs of patients. It's time to leave behind the limitations of MABEL and NOAEL and adopt a forward-thinking approach that leverages the power of QSP.
If you'd like to learn how your team can begin using QSP for immunotherapy research, let's talk.
Discuss My Project