Dose Escalation and Stratification Designs in Early Oncology
Development
Summary Info
SAD (Single Ascending Dose) and MAD (Multiple Ascending Dose)
The SAD/MAD dose escalation strategy is crucial for:
Key Considerations in Dose Escalation
1. Is there a
range of safe doses where we can explore efficacy? - Yes, the
therapeutic window shown indicates a range of doses where the efficacy
is high and toxicity is minimal. This range allows for the exploration
of optimal dosing to maximize therapeutic benefits while minimizing
adverse effects.
The dose escalation process is crucial in clinical trials for several reasons:
Methodology of a dose escalation process in clinical trials.
Early Phase Decision-Making Stages
Decision Making Approaches
By employing adaptive designs, Bayesian methods, and biomarker-based endpoints, clinical trials can become more flexible and responsive to emerging data, potentially reducing development times and improving patient outcomes. These strategies also align well with modern regulatory frameworks that encourage innovation and efficiency in drug development.
Decision Framework
The evolution of cancer treatment strategies and trial designs has undergone significant transformations since the 1960s and 1970s. Initially, the predominant treatment for cancer involved chemotherapeutics, guided by the principle that the largest feasible dose was likely the most effective. This notion shaped early clinical trial designs, which were primarily based on establishing the maximum tolerated dose (MTD) of a drug, assuming that the highest dose that a patient could tolerate without severe toxicity would also be the most effective for treating cancer.
However, this assumption is no longer valid with the advent of modern oncology therapies, particularly targeted therapies and immunotherapies. These newer treatments have shifted the focus from MTD to identifying an optimal therapeutic dose that balances efficacy with minimal toxicity. This shift necessitates a different approach in clinical trial design.
Today’s oncology trials often utilize pharmacokinetics, pharmacodynamics, and biomarkers to determine the most effective dose. This is distinct from simply escalating doses until unacceptable toxicity levels are reached. The concept of a “therapeutic window” is now crucial, which represents a range where a drug is effective without causing significant side effects. This window is typically identified by analyzing the relationship between the probability of efficacy (represented by a green line) and the probability of toxicity (represented by a red line).
Modern trial designs also consider the dynamics of how drugs affect the body over time and how tumors respond to drugs. Unlike the past, where efficacy might be immediately apparent, newer treatments might show gradual improvements or require longer durations to assess true benefits, complicating the determination of when to judge efficacy or futility.
Additionally, the patient population for these trials is often selected based on specific biomarkers, which can predict a more favorable response to certain therapies. This biomarker-driven selection is part of a broader strategy of personalized medicine, which tailors treatments based on individual patient characteristics, potentially improving outcomes and reducing unnecessary exposure to ineffective treatments.
The challenges in contemporary oncology trials include:
In Details
Optimal Dose Not Based on Maximum Tolerated Dose:
Population May Be Defined by a Biomarker Signature:
Long Term Immune-Related Toxicity May Exist:
Traditional Efficacy Measures May Not Be Appropriate:
To tackle these complexities, adaptive trial designs are increasingly used. These designs allow modifications to the trial procedures based on interim data, which can include dose adjustments, sample size recalculations, or even changes in the study endpoint. Such adaptive designs are particularly useful in trials for combination therapies or for novel agents where the optimal therapeutic strategy is not yet well-defined.
3+3 design, a traditional method used in the dose-escalation phase of clinical oncology trials. This design has historically been a cornerstone in determining the maximum tolerated dose (MTD) but has encountered significant criticism for various limitations.
Historically, the 3+3 design has been a staple in oncology trials for determining the maximum tolerated dose (MTD). However, it has well-documented limitations such as the risk of choosing suboptimal doses for Phase 2 trials and the inability to efficiently include intermediate dose levels or handle varying cohort sizes (e.g., N=2, 4, 5).
These limitations highlight the need for more sophisticated designs in early-phase clinical trials, such as adaptive designs, Bayesian models, or model-informed drug development strategies. These approaches can offer more flexibility, utilize mathematical and statistical models to predict outcomes better, and are more responsive to data gathered during the trial. Such improvements could lead to more accurate dosing recommendations, reduced trial durations, and better overall outcomes for patients in clinical trials.
FDA Guidance on Dose Optimization: Recent FDA draft guidance and other materials focus on optimizing dosing strategies before large-scale trial phases. This includes recommendations for randomizing between doses to better establish efficacy and safety profiles without the constraints of traditional dose escalation methods.
Incorporation of Real-World Data and Biomarkers: There’s an increasing emphasis on integrating biomarkers and real-world data into trial designs. This approach helps in refining patient selection and enhancing the predictive accuracy of trial outcomes, facilitating personalized treatment strategies.
Late Toxicities and Safety-Efficacy Balancing: One of the persistent challenges in oncology drug development is managing late-onset toxicities. Research continues into how to incorporate safety and efficacy data into trial designs effectively, ensuring that treatments are both safe and beneficial over the long term.
Optimizing Combination Therapies: As oncology treatments increasingly involve combinations of drugs, figuring out how to dose each component effectively while managing interaction effects is a major focus area. This includes determining how to sequence drugs and adjust dosages based on patient response and toxicity.
The Guidance is designed to enhance the clinical trial process by focusing on optimizing dosing strategies before the commencement of large-scale approval trials.
Shift in paradigms for dose determination in oncology from the traditional Maximum Tolerated Dose (MTD) to the Minimally Active Dose (MAD) in the context of targeted immuno-oncology agents.
For Clinical Implications:
This transition in dosing strategy highlights a fundamental change in how newer oncologic therapies are developed and administered, emphasizing the need for a personalized approach to cancer treatment.
Exposure-Response Modeling in Clinical Development
Regulatory Emphasis on Dose-Finding Trials
Importance of Modeling and Simulation
Comprehensive Assessment: Beyond merely establishing safety and tolerability, this approach integrates the evaluation of how well the drug engages its target and the corresponding biomarker responses. This holistic assessment helps in making a scientifically grounded decision about the optimal therapeutic dose that should be carried forward into further clinical trials.
Flexibility in Treatment Options: By thoroughly understanding the dose-response relationship and having clear documentation, it becomes easier to make post-approval adjustments such as combining the drug with other therapies, modifying the treatment regimen, or altering the route of administration. These modifications can help optimize therapy based on real-world efficacy and safety data or new scientific findings.
Enhanced Confidence in Dosing Decisions: By the time a drug enters Phase 3 trials, having a well-characterized dose-response from earlier phases reduces the uncertainty and risk associated with efficacy and safety outcomes. This can lead to more efficient Phase 3 trials with a higher likelihood of successful outcomes.
Streamlined Regulatory Review: Comprehensive dose-finding studies provide robust data that can facilitate smoother regulatory review processes. Regulators are equipped with clear evidence supporting the chosen dosages, which can expedite approval timelines and decrease the likelihood of regulatory delays due to inadequate dosing data.
Overall, incorporating a dose-finding trial after initial dose escalation is crucial for refining dosage recommendations, ensuring regulatory compliance, and ultimately enhancing the drug’s clinical success by firmly grounding dosing decisions in scientific evidence. This approach is particularly important in the field of immuno-oncology, where the therapeutic window can vary significantly among patients and targeted mechanisms.
Note: Biomarkers assuming increasing importance in Phase 1 trials
The use of biomarkers in Phase 1 trials of immuno-oncology agents is increasingly pivotal. Data from a study encompassing trials from 2014 to 2020 shows that biomarkers are commonly measured in blood and significantly influence clinical development decisions, including dosage adjustments.
Biomarkers confirm that the drug is affecting its intended target (target engagement), enhancing the understanding of the drug’s mechanism of action. More than 50% of the studies reported that biomarkers had a direct impact on clinical decisions.
Recent regulatory changes emphasize the need for informative PD biomarkers in trial designs. These changes are expected to play a significant role in future clinical trial setups and drug development strategies.