As oncology treatments advance, optimising dose selection is crucial to improving patient outcomes. The FDA’s Project Optimus reshapes traditional dose-finding methods, prioritising both efficacy and long-term tolerability. This article covers Project Optimus’ background, objectives and implications for oncology trials.
As of early 2025, the FDA finalised guidance on dose optimisation under Project Optimus, reflecting an evolved regulatory approach. These documents emphasise the early integration of dose-ranging studies, incorporation of biomarkers and a transparent rationale for dose selection, now standard in oncology drug development.
Project Optimus is an initiative launched by the U.S. Food and Drug Administration (FDA) Oncology Centre of Excellence to reform dose selection and optimisation in oncology drug development. It responds to the evolution of cancer therapeutics, as targeted therapies and immunotherapies offer greater efficacy and tolerability than traditional cytotoxic agents.
Historically, oncology trials relied on the maximum tolerated dose (MTD). However, this approach is unsuitable for modern treatments with different dose–response relationships and wider therapeutic indices. Project Optimus promotes a more rational, ethical and scientifically grounded approach to dose selection.
The primary goal of Project Optimus is to choose doses that maximise efficacy while minimising toxicity, using dose–exposure, pharmacodynamic, toxicity and activity data to guide decisions. As cancer treatments advance, the FDA is modernising dose-selection practices. By moving away from outdated MTD models, Project Optimus advocates methods that account for targeted and immune-based therapies.
Project Optimus repositions oncology development away from traditional MTD-based designs, recognising that modern therapeutics often achieve optimal efficacy at below-MTD doses. The initiative calls for nuanced evaluation of dose–response, considering target engagement, biomarker changes and long-term tolerability.
A key aspect to this change of approach is the emphasis on conducting randomised, dose-ranging studies early in clinical development. These studies aim to characterise the relationship between dose, exposure, and both efficacy and safety outcomes. By doing so, developers can identify a dose or doses that offer the best balance of benefit and risk for patients.
Project Optimus stresses finding an optimal therapeutic window that maximises anti-tumour activity while minimising toxicity. Recognising that the highest tolerable dose may not be ideal for sustained use, it encourages evaluation of multiple doses for both short-term response and long-term outcomes like progression-free and overall survival.
Safety considerations extend beyond dose-limiting toxicities to include chronic low-grade adverse events affecting quality of life and treatment adherence, reflecting a patient-centric approach. Engaging patient advocacy groups in study design integrates patient perspectives on dosing frequency, administration route and daily-life impact.
Optimising doses is complex, due to cancer biology, patient variability and emerging modalities such as targeted therapies, immunotherapies and combinations.
Finding the optimal dose involves pharmacokinetics, pharmacodynamics, tumour biology, patient characteristics and treatment goals. A one-size-fits-all model rarely applies across cancer types, therapy lines or combinations.
Assessing long-term tolerability in early-phase trials is challenging. Project Optimus emphasises new approaches to data collection and analysis, and redefines tolerability metrics in oncology trials.
Regulators must evaluate benefit–risk across dosing regimens, balancing patient needs with detailed dose-optimisation data. Project Optimus highlights integrating biomarkers for target engagement and using endpoints beyond response rates, such as quality of life and durability of response.
Implementing Project Optimus may require new guidance tailored to dose optimisation. Patient-centric communication and well-designed studies can enhance enrolment and retention.
Project Optimus introduces expectations that reshape early-phase trials. Sponsors now require larger, longer Phase I clinical trials with detailed dose-ranging data before registrational studies, extending timelines, and increasing costs.
These demands can strain smaller biotech firms. Early strategic alignment with regulators and thorough planning are essential for compliance and efficiency.
To meet Project Optimus expectations, companies use advanced statistical techniques and modelling. Bayesian models and machine learning elucidate dose–response relationships and simulate trial scenarios. Bayesian adaptive designs allow real-time dose adjustments, reducing patient exposure to sub-optimal doses.
Integration of real-time data into adaptive clinical trials refines dosing strategies as trials progress, enhancing decision-making.
Industry response is mixed: stakeholders support patient-centric aims but cite longer timelines, complexity and cost pressures, especially for rare cancers.
Some companies now start dose-finding earlier and broaden cohorts for long-term tolerability. For example, a mid-sized biotech added two dosing cohorts with extended follow-up for exposure–response analysis. Another shifted from a 3+3 design to a model-based adaptive design with biomarker stratification, aligning with FDA expectations.
Project Optimus has commercial implications: detailed dose optimisation may favour HTA bodies and reimbursement. Early investment in optimisation can reduce post-approval safety issues and strengthen real-world outcomes. Cross-functional collaboration ensures alignment of regulatory, clinical, and commercial goals.
As oncology becomes more precise, ethical questions arise about balancing patient safety with rapid access to therapies. While randomised dose-ranging can improve outcomes, it may delay access to life-saving treatments. Sponsors explore hybrid or adaptive designs that allow expedited access while collecting comparative dose data.
There is an ethical imperative to avoid exposing patients to unnecessarily high doses. Incorporating bio-ethical frameworks and patient input ensures respect for autonomy and quality of life.
An often-overlooked aspect is the impact on access and equity. Increased complexity and costs may be passed to healthcare systems and patients. Trial access may worsen for underserved or resource-limited populations. Collaboration on decentralised trials, telehealth and community networks can mitigate disparities.
Optimal dosing requires new designs and methods based on data, incorporating adaptive approaches and real-time feedback.
Project Optimus recommends comparing multiple dosages before registrational trials to evaluate the therapeutic window comprehensively.
Adaptive Trial Designs
Adaptive designs, such as CRM and response-adaptive randomisation with Phase I/II transitions, allow dosage adjustments based on emerging data, improving optimisation efficiency.
Key techniques include advanced modelling, simulation and PK/PD analyses to guide dosing decisions based on data.
Population PK, exposure–response, PBPK and QSP modelling help map complex dosing landscapes.
PK/PD analyses establish relationships between exposure, target engagement and outcomes, identifying biomarker responses and patient variability.
Innovations enhance precision and personalisation, using biomarkers, real-world evidence and patient-centred strategies.
Although FDA-led, Project Optimus influences international regulators. EMA and PMDA monitor its implementation, and future harmonisation could standardise global dose-optimisation expectations. Aligning early with these evolving guidelines benefits developers worldwide.
Technological advances continue to reshape oncology dose optimisation, offering new avenues for personalisation, efficiency, and precision. Key innovations include:
Liquid Biopsies and ctDNA Analysis
Enables frequent monitoring and dynamic dose adjustments.
Artificial Intelligence (AI) and Machine Learning (ML)
Predicts individual dose–response patterns and informs selection.
Digital Health Technologies
Wearables and apps provide real-world tolerability insights.
In Silico Clinical Trials
Virtual cohorts test dosing scenarios rapidly.
Single-Cell Analysis
Reveals tumour heterogeneity to guide differential dosing.
As Project Optimus principles integrate into standard practice, oncology development will yield more effective, tolerable treatments, improving patient outcomes worldwide. Success will manifest in personalised dosing, fewer modifications, better adherence, and confidence in benefit–risk profiles.
To align with evolving expectations, sponsors gain a competitive advantage by adopting Project Optimus. Quanticate’s cross-functional expertise ensures your programme delivers optimised, evidence-based dosing that meets FDA and patient expectations. If you are looking for expert support in your oncology trials, please submit an RFI, and a member of our team will be in touch.
Bring your drugs to market with fast and reliable access to experts from one of the world’s largest global biometric Clinical Research Organizations.
© 2025 Quanticate