Abstract
Mathematical models have played a significant role in the development of current chemotherapy and radiotherapy treatment protocols. The widespread use of cytotoxic drugs has shaped the paradigm of uniformly administering a ‘maximum tolerated dose’ to patients; however, this approach fails to account for the dynamic and heterogeneous nature of challenging cancers, including metastatic disease. Recent clinical trials and regulatory decisions have aimed to address these issues by integrating mathematical models to drive preclinical experiments and personalize treatment schedules. By capturing mechanisms of dose-response dynamics, ecological dynamics like tumor-immune interactions or competition dynamics, and evolutionary dynamics across different therapeutic regimens, mathematical models hold the potential to advance current therapeutic strategies. As more preclinical and clinical data become available, the integration of mathematical models with ‘virtual patient’ frameworks, including ‘digital twins’, and artificial intelligence methods could further advance the mechanistic complexity and decision support capabilities of such models. Nonetheless, translating mechanistic models to routine clinical workflows will require overcoming current translational barriers, notably access to clinical data in standardized formats and regulatory constraints. Overall, recent trials demonstrate the promise of the field of mathematical oncology in translating predictive dynamics into treatment decision-making beyond the ‘maximum tolerated dose’ approach.
1. Introduction
Cancer is inherently complex, dynamic, and exhibits significant heterogeneity, both between and within patients, especially in metastatic settings (1–3). Differences are visible across multiple scales: from the genetic and epigenetic scale, over cells and tissues, and up to the organism level (2, 4). Across these scales, the growth of tumors and metastases, their interactions with the microenvironment, and their response to treatment exhibit complex and nonlinear spatial and temporal dynamics.
In this complex and dynamic setting, the current treatment paradigm of uniformly and continuously administering a ‘maximum tolerated dose’ (MTD), i.e. the largest possible dose that patients can tolerate with acceptable side effects, oftentimes fails as it leads to disease relapse due to the emergence of drug resistance (5). Furthermore, the MTD paradigm was developed during the ‘era of cytotoxic drugs’ (1, 6); however, most new cancer therapeutics today (such as targeted therapies or immunotherapies) have a different mode of action. Experience with current therapeutics shows that dose efficacy can saturate, which leads to additional toxicity without significantly enhancing efficacy (6). This effect is further exacerbated when evaluating drug combinations (1, 7). Determining optimal treatment strategies that reflect the complexity of treatment and its induced effects therefore remains a critical issue (6, 8).
‘Mathematical Oncology’ (2, 9–15) is a growing discipline in which mechanistic mathematical models are integrated with experimental and clinical data to improve clinical decision making in oncology. Such mathematical models are often based on biological first principles to capture spatial or temporal dynamics of the drug, tumor, and microenvironment. Models may be used to understand the complex and multi-scale nature of cancer, predict outcomes, and derive personalized treatment schedules. These approaches stand in contrast to recent machine learning and artificial intelligence methods, such as neural networks, which focus on mechanism-agnostic extraction of information from large-scale data (16).
In this review, we focus on how mathematical models are integrated within clinical workflows: influencing chemotherapeutic treatment scheduling, helping establish the MTD paradigm, and planning radiation treatments. We then present the latest pilot and phase I/II trials in which mathematical oncology is advancing patient care beyond the MTD paradigm by capturing treatment and eco-evolutionary dynamics and allowing for patient-specific treatment personalization. We believe that in the near future, tighter integration of models with novel computational tools (including virtual trials, digital twins, and artificial intelligence) will further advance translation in the field. This will, however, require overcoming current translational barriers, including the lack of standardized and accessible clinical data and regulatory constraints.
2. Mathematical Models Drive Novel Treatment Decisions
Mechanistic mathematical models use equations to represent the underlying processes within a system, rather than just inputs and outputs (2, 17). Most clinical cancer models describe quantities of interest over time, such as tumor size dynamics or drug concentrations (e.g., measurable in the plasma or delivered to the tumor). These models may capture treatment dynamics, including the dose-response of systemic drugs (18) or radiotherapy (19), and eco-evolutionary principles, such as ecological interactions of cell-based immunotherapies (20–22) or evolutionary dynamics due to the emergence of resistance (23). The results of these modeling approaches can inform treatment planning with respect to drug dosing, timing, and drug combinations. Recent trials are showing promise of adapting such mechanistic models into clinical pathways, which generally requires a workflow that integrates experimental and/or clinical data to derive treatment strategies (9, 15, 24), as portrayed in Figure 1.
Figure 1. Mathematical Oncology derives personalized treatment strategies.

Mechanistic mathematical models integrate experimental and/or retrospective clinical data to capture tumor, drug, or microenvironmental dynamics reflecting patient heterogeneity to infer personalized treatment strategies on a cohort or individual level. Inferred strategies may not only include drug sequencing, as depicted in the schematic, but may also evaluate dosing, combinations, etc. Model calibration might further require multiple iterations of generating hypotheses about the underlying mechanisms, and testing these with experimental or clinical evidence.
Constructing a mathematical model first requires making assumptions about the underlying biological processes at play, and how they interact to produce the observed dynamics. These hypotheses may be tested by calibrating mathematical models to available preclinical or clinical data. If the initial model assumptions do not capture the dynamics of the data, this can generate questions about the underlying biological mechanisms that lead to novel experimental or clinical studies aimed at answering them. A particular strength of mechanistic models is that they can be calibrated to capture heterogeneity and variability of dynamics across different scales (i.e., different tumors, patients, or cohorts), since various sets of model parameters can be chosen to reflect the corresponding heterogeneity. Although the limited amount of patient-specific data from an individual necessitates the use of less complex models compared to those that can be calibrated from experimental data sets, the use of modeling for individual personalized treatment has value in that it can incorporate expected heterogeneity based on retrospective clinical data and biological processes understood from preclinical data.
Once mathematical models have been calibrated satisfactorily with available data (15, 25, 26), they can be used to make predictions or generate treatment recommendations on a cohort or individual level. Using model predictions to derive effective treatment doses or schedules not only avoids a trial-and-error approach that would require excessive experimental and clinical resources but also bridges the divide between experiment and clinic by integrating a mechanistic understanding from various sources.
In the following, we highlight the clinical use of mathematical models historically and currently, capturing different aspects of drug and tumor dynamics. Figure 2 graphically represents specific modeling examples we discuss in this review (27–29). Starting with simple models of tumor growth curves and dose-response (Figure 2A–B), model dynamics have increased in complexity both in terms of tumor heterogeneity (Figure 2C) and drug delivery (Figure 2D). More recently, models have begun to embrace further the multi-scale nature of both tumor and non-tumor components (Figure 2E–F), as well as consider multi-drug treatment schedules. A separate overview of the latest modeling-informed clinical trials is also provided in Table 1, continued from (14). Overall, we will show that mathematical models can be used to drive clinical decisions that capture the complex, multi-scale, and dynamic mechanisms involved in cancer treatment.
Figure 2. Mathematical models capture various aspects of the underlying disease and treatment dynamics of cancer; ordered by model complexity with respect to the multi-scale nature of the disease.

(A) The Norton-Simon model describes tumor growth following a Gompertzian law, in which tumor cell kill depends on the proliferation rate of the tumor. (B) The linear-quadratic model displays differential dose-response curves for tissues with high (red) vs. low (blue) α/β ratios. (C) Adaptive therapy prolongs time to progression by individualizing treatment breaks that allow drug-sensitive cells to compete with resistant cells. (D) Pharmacokinetics and pharmacodynamics aim to bring drug concentrations within a therapeutic window (highlighted in gray) that balances efficacy and toxicity. (E) Extinction therapy aims to eradicate tumors via a first-strike-second-strike strategy. (F) CAR-T cell kinetics can be described to follow three phases: expansion, contraction, and persistence.
Table 1.
Overview of recent mathematical modeling-informed clinical trials.
| Model Type | Trial ID | Trial Name (abbreviated) | Trial Start | Trial Type | Est. #Patients | Outcome |
|---|---|---|---|---|---|---|
| Norton-Simon | NCT02595320 | Capecitabine in Metastatic Breast and GI Cancers (X7–7) | 2015 | Phase 2 | 200 | Reduced toxicity ✓ |
| NCT02650752 | Intermittent High-Dose Lapatinib in Tandem With Capecitabine for HER2 Overexpressed/Amplified Metastatic Breast Cancer With CNS Metastases | 2016 | Phase 1 | 11 | Feasibility and Efficacy ✓ | |
|
| ||||||
|
Dynamics-based
Radiotherapy |
NCT03557372 | Mathematical Model-Adapted Radiation In Glioblastoma | 2018 | Phase 1 | 14 | Feasibility and Safety ✓ |
| NCT03768856 | Temporally Feathered Radiation Therapy (TFRT) for Head and Neck Squamous Cell Carcinoma | 2018 | R-IDEAL Stage 1/2A |
5 | Feasibility and Toxicity Assessment ✓ |
|
| NCT03656133 | PSI for personalized radiotherapy of HPV+ Oropharyngeal Cancers | 2018 | Phase 2 | 55 | Improve Rapid Response ✓ | |
|
| ||||||
|
Evolution-based:
Focus on PK/PD |
NCT01967095 | Low Dose Daily Erlotinib in Combination With High Dose Twice Weekly Erlotinib in Patients With EGFR-Mutant Lung Cancer | 2013 | Phase 1 | 53 | Tolerability ✓ |
| NCT03810807 | Dacomitinib and Osimertinib for Patients With Advanced EGFR Mutant Lung Cancer | 2019 | Phase 1 | 22 | Active, not recruiting | |
|
| ||||||
|
Evolution-based:
Adaptive Therapy |
NCT02415621 | Adaptive Abiraterone Therapy for Metastatic Castration Resistant Prostate Cancer | 2015 | Early Phase 1 | 19 | Active, not recruiting |
| NCT03630120 | Adaptive TKI Therapy for Thyroid Cancer | 2018 | Phase 2 | 6 | Terminated, due to Lack of Efficacy |
|
| NCT03511196 | Intermittent Androgen Deprivation Therapy for Stage IV Castration Sensitive Prostate Cancer | 2018 | Early Phase 1 | 17 | Active, not recruiting | |
| NCT03543969 | Adaptive BRAF-MEK Inhibitor Therapy for Advanced BRAF Mutant Melanoma | 2018 | Early Phase 1 | 14 | Active, not recruiting | |
| NCT05393791 | Adaptive vs. Continuous Abiraterone or eNZalutamide in mCRPC (ANZadapt) | 2022 | Phase 2 | 168 | Recruiting | |
| NCT05080556 | Adaptive ChemoTherapy for Ovarian Cancer (ACTOv) | 2023 | Phase 2 | 80 | Recruiting | |
| NCT05651828 | Adaptive Therapy of Vismodegib in Advanced Basal Cell Carcinoma | 2023 | Early Phase 1 | 34 | Recruiting | |
| NCT06734130 | Adaptive Androgen Deprivation and Docetaxel in Metastatic Castration Sensitive Prostate Cancer | 2025 | Phase 2 | 25 | Recruiting | |
|
| ||||||
|
Evolution-based:
Extinction Therapy |
NCT04388839 | Evolutionary Therapy for Rhabdomyosarcoma | 2020 | Phase 2 | 28 | Recruiting |
| NCT05189457 | First Strike, Second Strike Therapies for mCSPC | 2022 | Phase 2 | 32 | Active, not recruiting | |
| NCT06409390 | Sequential Therapies Modeled on Evolutionary Dynamics for Breast Cancer | 2024 | Early Phase 1 | 15 | Recruiting | |
|
| ||||||
|
Fully
Personalized Treatment |
NCT04343365 | Evolutionary Tumor Board (ETB) | 2020 | Observational | 35 | Recruiting |
| NCT06423950 | Multicenter, Pediatric and AYA Evolutionary Tumor Board (pedsETB) | 2024 | Observational | 35 | Recruiting | |
Abbreviations: Tyrosine Kinase Inhibitor (TKI), mCSPC: Metastatic Castration Sensitive Prostate Cancer, mCRPC: Metastatic Castration Resistant Prostate Cancer, CNS: Central Nervous System, ✓: Successfully evaluated
2.1. Historical Beginnings in Chemotherapy: Log-Kill & Norton-Simon Models
Historically, mathematical models in oncology described simple tumor growth and dose-response dynamics of chemotherapeutic treatment, initially in leukemia and breast cancer. In the 1960s, Skipper et al. (30) developed the log-kill model, which revolutionized the treatment of childhood leukemia and helped establish many critical concepts such as multicycle and combination chemotherapies, as well as today’s MTD paradigm (5, 31–33). Using data from a murine model of leukemia, they described tumor cell growth using an exponential / logarithmic growth law, i.e. tumor cells doubling at a constant rate until reaching a lethal threshold size. This also implied fractional killing of cells relative to dose; if a single treatment dose reduces tumor burden e.g. from 109 to 108 cells, the same dose reduces a burden of 105 to 104 cells. Due to such diminishing returns in absolute cell kill, this model suggested that chemotherapeutic agents should be given as frequently as possible (e.g., in combinations or when patients exhibit minimal residual disease), which challenged the prevailing paradigm of the time (5, 32). Freireich and Frei (34) then hypothesized that the combination of chemotherapies would be additive under the log-kill model (35), leading to the development of the four-drug VAMP regimen (36) for pediatric acute lymphocytic leukemia (ALL). Despite initial reticence (35), the concept was highly successful and evolved into a series of combination therapies that raised the 10-year survival of pediatric ALL patients from 10% to over 90% (35, 37).
Norton and Simon (27) further refined the log-kill model in the late 1970s to describe the growth of solid tumors and their response to treatment (5, 32, 33, 38). At this time, the convention for chemotherapy dose was that intensity should be lowered after patients achieved complete remission to manage the cumulative toxicity or prevent secondary malignancies (27). In their landmark study (27), Norton and Simon challenged this convention by using mathematical models to derive insights from experimental data of various tumors that matched clinical experience. They fit untreated growth to a Gompertzian growth law, which represents exponential growth with a steadily decaying growth rate; the drug-induced death rate depended on the proliferating fraction (as shown in Figure 2A) due to the antimitotic mechanism induced by chemotherapy (27). The model implied that small residual tumors required much larger doses than previously assumed due to their large growth rates and suggested that treatment breaks should be shortened to avoid tumor regrowth (39). This dose-dense approach was then validated in a phase III trial of adjuvant chemotherapy in breast cancer (40), significantly improving clinical outcomes without increasing toxicity, and is currently the preferred dosing schedule for HER2-negative breast cancer patients receiving adjuvant chemotherapy (41).
Traina et al. (42) later extended the Norton-Simon model to improve chemotherapy dosing schedules of capecitabine for breast cancer, which comes with significant dose-limiting toxicity. They modeled tumor xenograft response dynamics with a similar Gompertzian model under the conventional capecitabine schedule and found that treatment efficacy decreased within the administration window of the conventional schedule. Subsequently, they identified in vivo that shortening the administration time of the drug could improve survival (42). This novel schedule has since been tested in various phase I/II trials (42–46), both as monotherapy and in combination, and a recently completed phase II trial (47) found that the novel dosing schedule resulted in less toxicity while retaining similar efficacy.
Crucially, these relatively simple models continue to have a significant impact on today’s treatment paradigm and first demonstrated the capability of mathematical models to infer novel treatment schedules informed by experimental or clinical evidence.
2.2. Modeling & Personalization in Radiation Oncology
Mathematical modeling has also greatly contributed to advancing radiation therapy treatment scheduling by describing crucial dose-response dynamics (3, 19, 28, 32). The linear-quadratic (LQ) model forms the basis of radiotherapy planning and optimization in the clinic by quantifying dose-responses in the tumor or surrounding normal tissue (19, 28, 32, 48, 49). It describes the survival probability S of a cell being exposed to a radiation dose D by S = , where α and β reflect the cell’s radiosensitivity. Intuitively, the model is often described as inducing cell death from single or multiple radiation ‘hits’ via the α and β parameters (28), respectively, which can be interpreted as an extension of the log-kill model.
Douglas and Fowler first quantified the effect of radiation dose fractions using the LQ model in the late 1970s (28, 50), fitting to in vitro and in vivo data. During the late 1980s, it was then identified that tumors with high proliferation rates should exhibit a dose-response behavior with higher α/β ratios than the surrounding healthy tissue (28, 32, 51), as exemplified in Figure 2B (28). This implied that it would be beneficial to divide a total dose into smaller but more frequent fractions, i.e. using a hyperfractionation schedule, as it maximizes tumor cell kill while minimizing damage to healthy tissue. Such a benefit of hyperfractionation was e.g. observed in head and neck cancer (32, 52), as it exhibits a high α/β ratio compared to other cancers (28, 32). Today, the LQ model has widespread applicability in the clinic (28, 49), and the model’s simplicity has been attributed as a major contributing factor to its successful translation.
Beyond predicting tissue-specific radiation response, mathematical modeling may also capture the inherent heterogeneity of tumors (3, 19). Leder et al. (53) employed the LQ model to consider the dose-response of PDGF-driven glioblastoma and iteratively refined a mechanistic cell-population model (incorporating temporal dynamics of sensitive and resistant cell growth) through comparison of predicted tumor outcomes with murine experiments. Based on this successful model calibration, they derived a novel treatment schedule that showed prolonged survival over conventional schedules in vivo. The feasibility and safety of this computationally-derived schedule were subsequently trialed in a recent phase I study (54), exemplifying the potential of more complex mathematical approaches to derive novel treatment schedules.
The LQ model may also be extended to account for tissue recovery after radiation, or in combination with genomic data to bridge the multi-scale nature of the disease (3, 19). López-Alfonso et al. (55) developed a nonlinear model of normal tissue recovery that suggested altering the fractional doses delivered to organs at risk to reduce normal tissue toxicity, where the feasibility of this approach was confirmed by a phase I study of head and neck squamous cell carcinoma (56). Furthermore, Scott et al. (57) combined a gene expression-based radiosensitivity index (58) with the linear-quadratic model to develop the genomic-adjusted radiation dose (3), which is being tested in a phase II trial (59), exemplifying the potential of integrating individual-specific molecular data with mathematical modeling.
A recently completed single-center nonrandomized phase II single-arm trial has also shown the potential of capturing patient-specific tumor heterogeneity to make individualized treatment decisions. The proliferation saturation index (PSI) (60) is a dynamic biomarker that estimates the proportion of proliferating radiosensitive cells, which can be estimated pre-treatment based on two conventional longitudinal tumor volume measurements using a logistic growth model (3, 19). Prokopiou et al. (60) proposed and validated the model through retrospective longitudinal data and showed that pre-treatment PSI was prognostic for radiation response. Furthermore, they identified in silico that stratifying patients into conventional and hyperfractionated radiation protocols based on pre-treatment PSI could improve the tumor response (60, 61). This was successfully validated in a prospective phase II trial for HPV+ oropharyngeal cancers (61, 62), increasing the percentage of patients who achieved a robust mid-treatment response (tumor volume reduction of more than 32% by week 4 of radiotherapy) from 50% to 58%, and showed that patient-specific tumor dynamics can be leveraged to personalize radiotherapy and improve responses.
Overall, recent trials exemplify that dose-response modeling (based on the LQ model) can be integrated with more complex disease scales (such as genomic information or individual-specific cell growth dynamics), which show promise to improve patient outcomes.
2.3. Modeling Drug Dynamics
Pharmacokinetic and pharmacodynamic mathematical models are used extensively for monitoring and predicting drug concentrations and their effects, with applications ranging from preclinical drug development to clinical decision-making. Pharmacokinetic (PK) approaches focus on the drug concentration profiles in the body, including uptake, distribution, metabolism, and clearance, whilst pharmacodynamic (PD) methods investigate the dynamic effects on the body and disease. Quantitative Systems Pharmacology (QSP) approaches extend these mathematical models to integrate drug dynamics across multiple scales (63–65) and are already aiding the design of clinical studies across all trial phases (64).
Many oncological drugs do not make it into clinical use due to high toxicity, complex pharmacology, and high variability between individuals (66). In these scenarios, the PK/PD dose-response relationship is crucial for identifying a therapeutic window (depicted in Figure 2D) where a drug has a high enough dose for an efficacious response, whilst constraining for severe dose-limiting, irreversible, or life-threatening toxic effects (18). The PK/PD time course may be nonlinear and/or multi-scale, as different complexities are considered in relation to the drug and system compartments to determine pharmacological, physiological, and pathological responses. Models are often individualized to patients and leverage prior knowledge using so-called nonlinear mixed-effects models, population PK models, or Bayesian methods (67). These models are usually implemented with the goals of maintaining a certain target exposure and personalizing treatment dose for efficacy and toxicity.
Maintaining a target exposure is a critical application for PK/PD models, as a fixed standard dose over a patient cohort may easily overdose or underdose individual patients. If the drug exposure is predicted to be suboptimal, adjustments may be made through escalation or de-escalation (dose changes), or intensification (frequency changes) guided by PK/PD models for dose optimization. Oftentimes, the goal is to achieve a particular target dose or a target total dose over time, denoted as area under the curve (AUC) (68–72). In an early study by Calvert et al. (68), dosing was adjusted to meet a target AUC for carboplatin using PK/PD models that considered peripheral and/or central drug concentrations separately. Model insights revealed that adjusting for body size was not as important as individualizing for variable renal clearance rates. In a later study in children with acute lymphoblastic leukemia by Evans et al. (69), dosing strategies personalized to individual PK clearance rates had a higher rate of continuous complete remissions compared to standard chemotherapy regimens based on body surface area.
Several studies also showed that dose personalization may be critical for keeping the measurable drug concentration within a range that balances efficacy and toxicity (73–75). Gamelin et al. (74) conducted a prospective multicenter phase III randomized trial comparing a standard body-weight constant dosing with individualized, PK-guided dose-intensified treatment of fluorouracil in metastatic colorectal cancer patients. They found a wide pharmacokinetic variability between patients and a large distribution of optimal doses that achieve the same plasma ranges in the personalized arm, which ultimately had a higher objective response rate (74). Furthermore, Long-Boyle et al. (76) developed a model calibrated with retrospective data, implementing a clinical tool to recommend busulfan dose to pediatric and young adult patients undergoing hematopoietic cell transplants. The model quickly reaches a steady-state target concentration depending on body weight, age, and clearance rates without exceeding toxic thresholds (76).
In certain cases, toxicity needs to be more explicitly modeled to monitor and predict dose limitations accurately, as there may be differences between patients due to age, body size, sex, or other PK factors. In the early 2000s, investigations on dose-densifying the administration of the standard first-line regimen of docetaxel plus epirubicin for metastatic breast cancer showed promising responses, but the more frequent administration was limited by the risk of severe, life-threatening neutropenia (77). PK/PD models were subsequently designed to enable personalized treatment of this drug combination, explicitly modeling drug pharmacokinetics, tumor size dynamics, and crucially, neutrophil counts, which guided intensification in a phase I/II trial (77, 78). These studies showed that unacceptable toxicities from densified regimens could be circumvented using model-driven drug administration. Moreover, PK/PD models have been developed and applied to achieve target drug clearance rates to balance efficacy and toxicity for docetaxel explicitly (79) and to personalize computational metronomic schedules of vinorelbine with non-small cell lung cancer and mesothelioma patients (80, 81). Specifically, children, the elderly, and patients with renal or liver impairment may have reduced drug clearance and higher toxicities (18, 66).
2.4. Evolution-based Treatment Strategies
Despite significant progress in drug development, metastatic and advanced-stage cancers remain largely incurable, since initial response is often followed by disease relapse over multiple lines of treatment (5, 23). Evolution of treatment resistance is driven by competitive interactions between cancer cells and complex interaction dynamics within the microenvironment and with other non-malignant cells (23). Mathematical modeling is particularly suited to capture such complex nonlinear dynamics, which has resulted in a rich history of numerous model-based evolutionary treatment strategies.
Early History of Modeling Resistance
Building on the Norton–Simon model discussed above, one of the first mathematical approaches to consider the emergence of treatment resistance from the 1980s is the Goldie–Coldman model. This model assumed that resistant cells originate from sensitive ones, due to spontaneous mutations occurring and accumulating prior to treatment. Logic follows that any delay in treatment would result in larger, more resistant tumors that are more likely to relapse (82, 83). However, this assumption is not universally true. The Goldie–Coldman model’s emphasis on early treatment aligned partially with pancreatic (84), colorectal (85), and breast (86, 87) cancer data. However, patient outcomes have since been observed to be independent of the timing of adjuvant chemotherapy across multiple cancer types, including in esophageal (88) and lung (89, 90) cancer studies. This strategy also argued for alternating rather than sequential application of multiple drugs (91), where cycles of each drug would be interleaved; however, clinical trials in breast cancer based on this mathematical model observed longer overall survival on the sequential arm, where each drug was given to completion before moving to the next (92, 93).
The potential of treatment breaks was identified by developing models to account for drug sensitivity of the tumor during the emergence of drug resistance using in vivo studies of hormone therapy in a mouse model of androgen-sensitive prostate cancer (94, 95). Intermittent approaches that switch between periods of the drug being on and off have since been tested prospectively in phase II clinical studies in locally-advanced prostate cancer, demonstrating reduced side effects and enhanced disease control relative to continuous therapy (96–98). Both continuous (99) and intermittent (100) drug schedules have been informed by mathematical modeling of the treatment response, characterizing the relative impacts of therapy on cellular turnover, mutation, and receptor regulation to explain the treatment responses observed clinically. Using models to predict tumor dynamics (101) and individual patient outcomes (102) also improved upon the prospective studies in the clinic that did not use modeling to create intermittent schedules. Mathematical modeling additionally enabled in silico comparison of different scheduling approaches (103) and the application of control theory to develop optimized intermittent treatment protocols (104).
Subsequently, novel competition-based models were developed as an alternative description of the emergence of treatment resistance during cancer therapy. These models assume distinct populations of drug-sensitive and -resistant cells, which, in contrast to the Goldie–Coldman model, compete for some shared resource. Shimada et al. (105) modeled prostate tumor dynamics assuming distinct, competing populations of drug-sensitive and resistant cells, demonstrating that the potential benefits of intermittent therapy are retained under the assumption of distinct sensitive and resistant populations. These mathematical models redefined the role of sensitive cells; their continued, intratumoral presence enabled by intermittent treatment holidays not only maintains the drug sensitivity of the tumor but also suppresses the growth of resistant cells through intercellular competition.
Personalizing Competition-Based Models: Adaptive Therapy
Selection and competition motivated Gatenby et al.’s work to personalize intermittent therapy schedules in the form of adaptive therapy (AT) (106). AT differs from intermittent treatment in that the timing of holidays is adapted to patient-specific dynamics (Figure 2C) rather than using fixed timing or burden thresholds across the cohort. The first clinical trial of AT was conducted in metastatic castration-resistant prostate cancer, where mathematical models guided the cycling of abiraterone therapy based on patient-specific prostate-specific antigen (PSA) levels (107). The primary adaptive protocol, inspired by model simulations, administered abiraterone and prednisone until a patient’s PSA level dropped by 50% from baseline, before suspending treatment until PSA rebounded above the baseline. The median time until disease progression more than doubled compared to a matched contemporaneous cohort on continuous therapy (33.5 months vs. 14.3 months) while drug exposure and associated costs were concurrently reduced (108). These results also displayed significant heterogeneity in patients’ responses to AT, motivating modeling efforts to identify the patient-specific benefit of AT over conventional treatment approaches (109, 110). Such mathematical approaches have since been extended to predict the expected benefit of AT after the first treatment cycle (bioRxiv 2025.04.03.646615), potentially enabling future clinical stratification of patients based on their expected response.
Clinical trials based on adaptive strategies for tumor control have been extended to metastatic castrate-sensitive prostate cancer (NCT03511196, NCT06734130), melanoma (NCT03543969), and basal cell carcinoma (NCT05651828). Further phase II AT trials in prostate (NCT05393791) and ovarian cancer (NCT05080556) also exemplify differing approaches to implementing AT, testing treatment break and de-escalation approaches, respectively. Recently published abstracts credit the ongoing role that mathematical modeling is playing in the design of current AT trials for melanoma (111) and breast cancer (112).
Combining Evolutionary Dynamics with Drug Dynamics
Whilst mathematical modeling for clinical use generally favors simplicity, drug dynamic models can be combined with evolutionary modeling to account for several factors at once. More recent PK/PD models have explicitly accounted for the heterogeneity of the tumor population, evolution of resistance, and its effect on optimal drug dosing (113–115). PK/PD models have been combined with simple tumor dynamic models, such as tumor-growth-inhibition models, which account for reduced drug response via increased drug resistance over time (113) and with Lotka-Volterra two-compartment models that represent drug-sensitive and drug-resistant cells (NCT05651828).
In another approach, Chmielecki et al. (116) built on a multi-type branching process model of sensitive and resistant cells (117, 118) and proposed that intermittent high-dose pulses combined with a lower daily dose of EGFR inhibitors could delay the emergence of resistance. In vitro cell culture experiments were coupled with a stochastic mathematical model that was calibrated with clinical pharmacokinetic data. The same group then extended the model to incorporate dose-dependent mutations of sensitive cells and showed that a pulsing strategy is effective irrespective of the dependency of the mutation rate on the drug concentration (114). This high-dose/low-dose pulsing approach was tested with erlotinib in a phase I clinical trial (NCT01967095), and while the strategy did not effectively delay the emergence of resistance, it was associated with fewer metastases (119). Poels et al. (115) later combined the same branching process model (118) with a PK model of osimertinib-dacomitinib combination therapy (120). Using an optimized dosing schedule to minimize tumor burden and toxicity, they observed a response rate of 73% in a phase I trial (NCT03810807), with fewer adverse events than osimertinib or dacomitinib monotherapies.
Targeting Tumor Heterogeneity via Combination Therapies
Combination therapies are emerging as a promising strategy to address tumor heterogeneity and resistance. In many hematologic malignancies, drugs have additive effects within single patients that yield potent cell kill by combining therapies (121), as we have e.g. seen at the beginning of this review with the VAMP regimen that exhibited additivity of log-kills in ALL. Statistical models including Loewe Additivity (122) or the Bliss Independence model (123) can account for intra-tumoral variability of response across different therapeutics. The Bliss Independence model assumes cells respond heterogeneously to independent drugs and converges to log-kill additivity for cancer chemotherapy (35). This model serves as a null model demonstrating synergy of drugs when the effect of the combination exceeds the expected response under independence (121). When the resistance to multiple drugs is due to independent mechanisms, the likelihood of cross resistance is successively lower with each additional drug (35, 124).
Solid tumors, in contrast, typically exhibit greater inter-patient heterogeneity in drug response and display a higher likelihood of pre-existing resistance (121, 124). Models focused on genetic mechanisms of resistance found that combination therapies would not succeed if single mutations could confer cross-resistance across all used drugs (124). Pioneering work by Palmer and Sorger (125) further demonstrated that the benefit of combination therapies across various clinical trials oftentimes can be explained by the independent action of drugs (i.e., low cross-resistance), with one therapy acting as the primary contributor to a patient’s response to combination therapy. Their methodology relied on a statistical model that was further refined by relying on the additivity of progression-free survival (PFS) times (126). By selecting published phase 3 combination trial results between 2014 and 2018 with available Kaplan-Meier PFS survival curves of the ‘standard of care’ therapy and of a single-agent therapeutic to be added, they were able to correctly predict combination trial PFS outcomes with 100% sensitivity and 78% specificity.
These findings have the potential to directly inform the design of combination therapy in future prospective trials (121), as computational tools can be used to predict the results of combination therapy trials before they begin or conclude. Furthermore, high single-agent activity implies that predictive biomarkers should be developed to personalize therapy to the most effective drug(s) for any individual patient (121). The search for such suitable biomarkers could also be supported by mathematical models (127).
Recent Evolutionary Therapies
Inspired by ecological principles driving mass-extinction events, an emerging form of evolution-based therapy is ‘extinction therapy’ (5), where mathematical modeling has suggested that tumor eradication could be achieved by administering treatments in a ‘first-strike-second-strike’ fashion (128). In this scenario, a ‘first strike’ would dramatically reduce tumor mass and heterogeneity via a first treatment, rendering the small remaining tumor population vulnerable to a ‘second strike’ — a sequence of secondary treatments which eradicate the remaining susceptible population (5, 128), as illustrated in Figure 2E. Extinction therapy is currently being evaluated in rhabdomyosarcoma (129) (NCT04388839), metastatic castration sensitive prostate cancer (NCT05189457), metastatic breast cancer (NCT06409390), and an extinction trial for Ewing sarcoma will be opening soon (130).
Acquiring resistance is an inherently dynamic process, and treatment personalization based on longitudinal monitoring is also starting to show great promise in counteracting disease recurrence (5, 131). Ongoing trials are investigating the feasibility of implementing real-time, model-informed decision support for individual patients with the Evolutionary Tumor Board (ETB) pilot studies, which integrate mechanistic modeling and evolutionary theory into the traditional tumor board concept (NCT04343365, NCT06423950) (132). In these studies, a multidisciplinary team that includes oncologists, radiologists, mathematicians, and evolutionary biologists uses patient-specific mathematical models to simulate tumor dynamics, forecast treatment outcomes, and guide future treatments. These models incorporate the evolution of resistance and possible treatment resensitization and are calibrated using longitudinal tumor burden data from previous treatment histories. This approach can guide what or why a specific treatment should be administered, and when, providing patient-specific decision support for the treating physician.
2.5. Immunotherapy
In recent years, numerous immunotherapy approaches have revolutionized treatment for various cancers. Novel immunotherapies, such as immune checkpoint inhibitors or Chimeric Antigen Receptor (CAR) T-cell therapy, are known to induce complex temporal and spatially heterogeneous effects (133), and thus, the dose-response relationships needed for immunotherapy modeling are often different from those developed for anti-proliferative and targeted agents. Toxicity considerations also differ. The limitations of traditional dose selection principles and trial designs for immunotherapy, based on the MTD paradigm, have been recognized (7, 134), and mathematical model-informed designs have been proposed (7).
The most notable and clinically successful model applications in immuno-oncology are the post-approval drug label changes of PD-1/PD-L1 inhibitors, based on computational modeling that supported e.g. the extension of dosing intervals (31, 113). The application of a PK model to data from phase I and phase III trials of atezolizumab (a PD-L1 inhibitor) to determine exposure-response relationships (i.e., considering both efficacy and drug-related toxicity) revealed that administering adjusted doses every two or four weeks would have comparable efficacy and safety as the previously approved three-week interval (135). This work, along with similar modeling for nivolumab (136) and others, supported the interchangeable use of different treatment schedules, offering patients and their healthcare providers greater flexibility without having to incur additional trials (113, 135).
Mathematical modeling has also played a role in regulatory approval of immunotherapies, with the FDA employing PK modeling to evaluate the first approved CAR-T cell therapy (tisagenlecleucel) (134). This model describes the expansion of effector cells after T-cell administration, and their subsequent conversion into memory cells, recapitulating the observed phases of patient T-cell dynamics (shown in Figure 2F) from the first CAR-T cell trials (29). The model also accounted for the effects of co-medications, in order to understand adverse events such as cytokine release syndrome (CRS) (134, 137), and remained crucial for the evaluation of subsequent CAR-T cell therapies (134).
As our mechanistic understanding of effective immunotherapy increases, the number of mathematical models that consider its multi-scale effects will also increase (64, 134, 138). Numerous mathematical modeling studies have considered tumor-immune interactions and drug dynamics in the last three decades (139, 140). The application of such models to identify promising combination therapies is also becoming more prevalent in industry (7, 140), as various ‘what-if’ scenarios targeting different parts of the larger cancer-immunity cycle (7) can be tested in silico. Overall, the approval of increasingly complex immune therapies (including combinations) requires a quantitative and mechanistic understanding of the underlying dynamics and therefore is an area where mathematical modeling will become vital.
3. Computational Tools Beyond Mathematical Modeling
We have shown here how mathematical models using equations that describe cancer and treatment dynamics can drive clinical decisions through prediction and decision support at the bedside. Crucially, translation of mathematical models to the clinic requires calibration via integration of experimental and clinical data. With the increasing availability of large troves of experimental and clinical data, novel computational methods can be used to assist with clinical translation advancement (141–143). We highlight here how virtual cohorts and artificial intelligence methods can be integrated into the workflow of mathematical model-driven decision support.
3.1. Mechanistic Virtual Patient Modeling
Understanding both inter- and intra-patient variability is a key application of modeling approaches in the clinical setting. One tool to capture this variability is a ‘virtual patient’ cohort (26, 144, 145). Individual virtual patients are in silico instances of an underlying model, calibrated from plausible ranges of biological and clinical data. When analyzed as a group, their simulated outcomes should reflect the heterogeneity observed in equivalent clinical cohorts. These virtual patients can be created from a combination of statistical models (e.g., sampling from survival models, Bayesian methods), machine learning and AI models (e.g., generative models, predictors conditioned on patient features), and mechanistic mathematical models (e.g., ODE systems, PKPD/QSP frameworks). Here we focus on mechanistic models, which are derived from the underlying biology of the cancer, calibrated with real-world data, and framed in a clinical context.
Many of the previously presented clinically-actionable models used virtual cohorts in some capacity to facilitate model translation (60, 115, 132). Prokopiu et al. (60) generated virtual patient cohorts to quantify the inherent uncertainty of parameter estimates for the dynamics-informed pre-treatment PSI biomarker for radiation response. Furthermore, Poels et al. (115) used an in silico trial approach to estimate the best combination therapy in a virtual population with inter-subject variability in pharmacokinetics and pre-existing resistance. The virtual cohort methodology was also pivotal in the studies led by Sorger and Palmer (35, 125, 126), which predicted the efficacy of drugs in combination using retrospective single-drug outcome data.
In general, model parameters for mechanistic virtual cohorts are sampled from distributions representing the plausible variability in patient biology (e.g., tumor growth rates, treatment efficacy, drug resistance) to predict outcomes on the population level (26, 144). By calibrating parameter values to retrospective data (including tumor growth curves, biomarkers, and demographic data), a virtual patient cohort can be matched to observed metrics in a real patient population. We believe that virtual cohorts are crucial for model generalizability and successful translation, as they allow models to explicitly account for variability across different scales (i.e., variability across measurements, or heterogeneity in patient outcomes).
Virtual cohorts can further be used in a variety of clinical applications, for example, to test hypotheses, design and simulate clinical trials, and to personalize treatment. Running virtual trials (26, 146, 147) on virtual cohorts is a rapid and cost-effective way to connect experimental and clinical data (26) and to explore trial scenarios in advance, where a rich set of longitudinal data (e.g., imaging or blood-based biomarkers) will be essential for model calibration. Mathematical models can then be leveraged to explore novel treatment strategies under a variety of assumptions, enabling investigators to test counterfactual ‘what-if’ scenarios on these virtual cohorts before committing significant resources to a trial in humans (26).
Furthermore, the virtual patient approach can be applied to the individual, instead of focusing on cohort outcomes alone. This methodology, often termed ‘digital twin’ or ‘patient avatar’ (148–151), could forecast the disease trajectories of various treatment options, essentially allowing clinicians to simulate different interventions for a patient and allowing for dynamic updates during the course of therapy. Early work by the Swanson lab significantly advanced the field, showing how spatial patient-specific mechanistic models could be incorporated with routinely available longitudinal imaging data in brain cancer (152–154). Further, there has been significant interest from industry in integrating the virtual patient and virtual trial methodology in immuno-oncology to optimize combination therapies using mechanistic mathematical modeling (7, 138, 140). Our ongoing Evolutionary Tumor Board studies (132) also demonstrate the clinical feasibility of a virtual patient-based decision support tool, which can serve both as an advisor to, and analyst for, the treating oncologist.
3.2. Translational Opportunities in the Era of Artificial Intelligence
There has been considerable interest in integrating artificial intelligence (AI) methods into clinical workflows in recent years (155). Most clinical applications have centered around diagnostic applications of machine learning (ML) in radiology and pathology (16, 156), but beyond imaging, the use of AI for therapeutic decision making has remained challenging (156). The lack of mechanistic interpretability of ML models is considered one of the most critical barriers for clinical translation (155). Further, the ability to generalize models from preclinical datasets to real-world clinical scenarios persists as a major limitation, particularly due to the lack of access to patient-specific data, inherently heterogeneous patient populations, and the dynamically changing nature of the disease (155). Furthermore, deriving novel strategies, such as adaptive therapy, would be difficult for a machine learning algorithm based on retrospective data from continuous therapy alone.
Mechanistic learning (31, 142), wherein mechanistic mathematical models and AI methods complement each other, has emerged as a promising avenue to bridge this translational gap, as similarly noted for other medical applications (24, 148). Mechanistic mathematical models with few parameters succeed at capturing and predicting temporal or spatial dynamics of sparse datasets, while machine learning methods excel at capturing patterns from high-dimensional data. On their own, machine learning models might not be suitable to incorporate explicit biological mechanisms or patient-specific longitudinal dynamics when data availability is sparse (142, 157, 158).
When combined, deep learning models may be used for image analysis, extracting tumor information across multiple scales (e.g., identifying key molecular markers or estimating volumetric tumor information through image segmentation). These results could subsequently inform mechanistic mathematical models (141). Current work shows the potential of using the two methodologies in conjunction (141), e.g. to infer patient-specific spatial tumor distributions for radiotherapy planning (159, 160). Machine learning methods are also emerging as a promising avenue to learn underlying mathematical model equations directly from data, as demonstrated by e.g. the SINDy framework (161, 162) or physics-informed neural networks (PINNs) (163). Alternatively, deep reinforcement learning can be used to optimize treatment protocols in complex disease settings, as seen in adaptive therapy (164, 165).
4. Barriers to Translation
Despite promising results from current clinical trials and the prospect of integrating novel computational tools with mathematical models, several translational challenges remain. The clinical data available to calibrate models in real time is oftentimes sparse (e.g., tumor burden is typically measured only every few months by imaging), lacking in biological detail (e.g., the fraction of resistant cells in a tumor often cannot be directly measured), and is often measured across very different biological scales (e.g., a mutational analysis versus a tumor biopsy image versus a CT scan). Differences between humans and experimental systems also complicate deriving the response, toxicity, and feasible administration schedules via modeling (53, 54). Integration of clinical data into a model must also account for uncertainty in patient measurements (e.g., imaging variability, biomarker noise, and detection thresholds).
Furthermore, mathematical models developed by industry can suffer from overfitting (166), and validation remains difficult due to a general lack of transparency (64, 140); yet, these models often support novel drug development. There is a need for both data and model sharing, as well as the use of open-source platforms (127, arXiv:2509.13360) to allow for independent analysis (121, 167) of these approaches. This could not only help bridge the gap between academia and industry but also improve model predictions and consequently clinical model robustness and impact. Two additional translational barriers we will discuss further are the lack of clinical data in standardized and accessible formats, and the regulatory barriers that hinder clinical adaptation of both mechanistic and personalized models.
4.1. Data Integration and Standardization
Despite the abundance of retrospective patient data in cancer research, the lack of automated and standardized methods to convert raw clinical records into model-ready datasets persists as a critical bottleneck of model translation (11, 31, 168). Clinical data is often stored in highly variable and unstructured forms, limited by inconsistent date formats, non-numeric lab values, institution-specific terminology, and extended metadata frequently embedded with protected health information. These inconsistencies, coupled with the limitations on data sharing due to privacy concerns, hinder collaboration and data reuse across institutions (169). Whilst clinicians have embraced the Electronic Medical Records (EMR), data recording is a secondary priority to patient care and is not typically designed with computational analysis in mind (11, 169). To overcome these limitations, medical centers may implement automated pipelines for data collection, cleaning, and anonymization, based on the FAIR data principles — Findable, Accessible, Interoperable, and Reusable (168). These principles provide both technical and ethical guidelines for transforming disorganized clinical datasets into standardized, reusable research assets. Standardization frameworks, such as the Observational Medical Outcomes Partnership Common Data Model (OMOP-CDM) (170), are further being adapted into practice and could inform mathematical models to guide clinical decision-making (169).
4.2. Regulatory Barriers - Modeling & Personalization
Regulatory pathways that would allow mathematical and computational models to enter standard clinical workflows are currently limited. As the clinical trial methodology has for decades been based upon the MTD paradigm, drug tolerability remains the primary design constraint (instead of optimal dose efficacy) (6, 8), and trials are generally designed to find a therapeutic regimen that works best, on average, for a selected patient population (171). Within this framework, it remains difficult to integrate strategies derived from mathematical models into standard clinical workflows.
However, the current paradigm is changing. Regulatory agencies, including the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA), denote mathematical modeling approaches that support drug research and development as ‘Model-Informed Drug Development (MIDD)’ methods (134, 172), which the FDA expects will become standard within drug development pipelines (134). The aforementioned flaws of the current MTD paradigm have further been recognized by the FDA (6), and it is actively promoting dose optimization via its ‘Project Optimus’ program (6, 8, 173) that requires a mechanistic understanding of drug action. The ‘Fit-for-Purpose’ initiative and the ‘MIDD Paired Meeting Program’ were also established to promote quantitative tools for drug development and to improve communication between modelers and regulatory agencies (134, 174).
Due to the growing concerns and recognized limitations of animal testing in cancer (7, 175), the FDA has further highlighted the critical role that mathematical modeling will play in improving the translational capabilities of preclinical studies while reducing animal use (175). The average rate of translation from animal model to clinical cancer trial is below 8% (175, 176), largely due to unexpected safety or efficacy issues (175). Major challenges are also associated with testing immunotherapeutic strategies due to interspecies differences (7, 140, 175). In silico modeling and novel in vitro human-derived systems (such as organoids (177) and microphysiological systems) have thus been recognized as an opportunity to improve the predictive relevance of preclinical drug tests while saving costs (175).
The field will also need to address the notable difficulty of running clinical trials designed for treatment personalization under traditional trial regulations. The traditional approach continues to perform dose escalation in phase I trials, before evaluating safety and efficacy during phase II trials, and conducting randomized comparisons against the standard of care in phase III trials. However, this paradigm is slowly changing from a regulatory standpoint; adaptive (178) and Bayesian (179, 180) trial designs are becoming increasingly common, allowing for dynamic changes to enrollment, treatment, and endpoint analysis during trial execution, as compared to standard trials where the protocol is fixed. The long-running I-SPY 2 adaptive trial in breast cancer (181) exemplifies an innovative trial design that aims to accelerate the discovery of successful drugs while limiting patient exposure to inferior arms. Innovations include adaptive randomization, where the probability of enrollment in each arm is a function of the current predicted success rate; rapid drop-out and success rules, to promote the most promising therapies and quickly discontinue futile ones; and early signal detection based on early biomarkers (181, 182). Overall, the success of the I-SPY 2 study has demonstrated the feasibility of implementing complex trial protocols that would be needed to test model-informed trials. Although some longitudinal MRI measures were included in this study (181, 182), they were handled statistically as covariates; explicit mechanistic modeling of longitudinal data, using mathematical tools such as virtual cohorts, could provide a deeper layer of dynamic analysis for integration in future trial designs.
Nonetheless, overcoming any regulatory barriers will require collaboration between stakeholders of the translational process to bridge the gap between mathematical models and routine clinical workflows. From the scientific perspective, the integration of mathematical and computational tools must include input from healthcare providers, experimentalists, and modelers, as seen during the ‘Integrated Mathematical Oncology’ (IMO) workshop that has facilitated communication between these parties for more than 10 years (183). However, the ongoing discussion also needs to include industry partners, regulators, software developers, and lawmakers, who are crucial stakeholders within the larger translational process (113, 155).
5. Conclusion
Mathematical models in the clinic have been employed across a variety of different cancers, treatment modalities, and various scales (ranging from small-scale molecular pharmacokinetic dynamics to large-scale tumor and microenvironmental influences). Historically, these were simpler models initially focused on understanding dose-response dynamics. Skipper’s model assumed that the dose effect is log-kill and additive across different treatment regimens, implying that the reduction of tumor size under treatment is greatest when the dose is densified in the shortest tolerable time window. This assumption formed the basis of many subsequent dose-response and tumor growth models, including the linear-quadratic and Norton-Simon models, that have shaped the current treatment paradigm across different cancers and treatment modalities for many decades.
As more effective treatment interventions were developed, the range of curable cancers expanded, but metastatic disease has remained largely incurable (23). In challenging cases, it has now become apparent that treatment strategies need to reflect the complex and dynamic nature of cancer that encompasses multiple scales (reflecting tumor heterogeneity, immune interactions, evolution of resistance, etc.), beyond the reductionist view of cancer as a disease of the genes. As these complex dynamics are better understood and clinical data become more readily available, treatment strategies are being tailored towards individual patient dynamics, improving outcomes across different therapeutic modalities and strategies.
However, incorporating complex models for personalized treatment regimens will require access to more detailed data. Increased model complexity will need higher resolution experimental and aggregated clinical data for calibration, while personalized treatment regimens necessitate individual patient-specific clinical data collected in standardized and accessible formats. While virtual patient methodologies and machine learning methods could help support the integration of such data, adapting model-driven treatment strategies into routine clinical workflows will require overcoming crucial translational barriers and regulatory limitations.
Overall, mathematical models in oncology integrate experimental and clinical evidence to provide mechanistic insights into drug and disease dynamics, and infer valuable treatment strategies both on a cohort and individual level. They have played a key role in shaping current treatment strategies of chemotherapy and radiotherapy, and recent trial results are showing the immense potential of mathematical modeling to improve clinical workflows — bridging the divide between the current MTD paradigm and the complex, multi-scale, and dynamic nature of cancer.
Acknowledgements
The authors thank Heiko Enderling for feedback regarding the PSI model and the subsequent clinical study. All authors gratefully acknowledge funding by the NCI via the Cancer Systems Biology Consortium (CSBC) U54CA274507 and support from the Moffitt Center of Excellence for Evolutionary Therapy. K.G. also acknowledges funding from the EPSRC CDT in Sustainable Approaches to Biomedical Science: Responsible and Reproducible Research - SABS:R3 (EP/S024093/1).
Footnotes
Disclosure of Conflicts of Interest
The authors declare no potential conflicts of interest.
References
- 1.Smoragiewicz M, Adjei AA, Calvo E, Tabernero J, Marabelle A, Massard C, et al. Design and Conduct of Early Clinical Studies of Immunotherapy: Recommendations from the Task Force on Methodology for the Development of Innovative Cancer Therapies 2019 (MDICT). Clinical Cancer Research: An Official Journal of the American Association for Cancer Research 2020; 26:2461–5 [DOI] [PubMed] [Google Scholar]
- 2.Anderson ARA and Quaranta V. Integrative mathematical oncology. Nature Reviews Cancer 2008; 8:227–34 [DOI] [PubMed] [Google Scholar]
- 3.Caudell JJ, Torres-Roca JF, Gillies RJ, Enderling H, Kim S, Rishi A, et al. The Future of Personalised Radiotherapy for Head and Neck Cancer. The Lancet. Oncology 2017; 18:e266–e273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hanahan D Hallmarks of Cancer: New Dimensions. Cancer Discovery 2022; 12:31–46 [DOI] [PubMed] [Google Scholar]
- 5.Strobl M, Gallaher J, Robertson-Tessi M, West J, and Anderson A. Treatment of evolving cancers will require dynamic decision support. Annals of Oncology 2023; 34:867–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tracey A FDA’s Brian Booth: ‘We Need to Reconsider Our Approach to Dose Selection’. The Cancer Letter 2022; 48 [Google Scholar]
- 7.Morrissey KM, Yuraszeck TM, Li CC, Zhang Y, and Kasichayanula S. Immunotherapy and Novel Combinations in Oncology: Current Landscape, Challenges, and Opportunities. Clinical and Translational Science. 2016; 9:89–104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Araujo D, Greystoke A, Bates S, Bayle A, Calvo E, Castelo-Branco L, et al. Oncology Phase I Trial Design and Conduct: Time for a Change - MDICT Guidelines 2022. Annals of Oncology 2023; 34:48–60 [DOI] [PubMed] [Google Scholar]
- 9.Anderson ARA and Maini PK. Mathematical Oncology. Bulletin of Mathematical Biology 2018; 80:945–53 [DOI] [PubMed] [Google Scholar]
- 10.Gatenby RA and Maini PK. Mathematical oncology: Cancer summed up. Nature 2003; 421:321–1 [DOI] [PubMed] [Google Scholar]
- 11.Rockne RC, Hawkins-Daarud A, Swanson KR, Sluka JP, Glazier JA, Macklin P, et al. The 2019 mathematical oncology roadmap. Physical Biology 2019; 16:041005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Baldock AL, Rockne RC, Boone AD, Neal ML, Hawkins-Daarud A, Corwin DM, et al. From Patient-Specific Mathematical Neuro-Oncology to Precision Medicine. Frontiers in Oncology 2013; 3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Altrock PM, Liu LL, and Michor F. The mathematics of cancer: integrating quantitative models. Nature Reviews Cancer 2015; 15:730–45 [DOI] [PubMed] [Google Scholar]
- 14.McDonald TO, Cheng YC, Graser C, Nicol PB, Temko D, and Michor F. Computational approaches to modelling and optimizing cancer treatment. Nature Reviews Bioengineering 2023; 1:695–711 [Google Scholar]
- 15.Aherne NJ, Dhawan A, Scott JG, and Enderling H. Mathematical Oncology and It’s Application in Non Melanoma Skin Cancer – A Primer for Radiation Oncology Professionals. Oral Oncology 2020; 103:104473. [DOI] [PubMed] [Google Scholar]
- 16.Luchini C, Pea A, and Scarpa A. Artificial Intelligence in Oncology: Current Applications and Future Perspectives. British Journal of Cancer 2022; 126:4–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bravo RR, Baratchart E, West J, Schenck RO, Miller AK, Gallaher J, et al. Hybrid Automata Library: A Flexible Platform for Hybrid Modeling with Real-Time Visualization. PLOS Computational Biology 2020; 16:e1007635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Barbolosi D, Ciccolini J, Lacarelle B, Barlési F, and André N. Computational oncology — mathematical modelling of drug regimens for precision medicine. Nature Reviews Clinical Oncology 2016; 13:242–54 [DOI] [PubMed] [Google Scholar]
- 19.Enderling H, Alfonso JCL, Moros E, Caudell JJ, and Harrison LB. Integrating Mathematical Modeling into the Roadmap for Personalized Adaptive Radiation Therapy. Trends in Cancer 2019; 5:467–74 [DOI] [PubMed] [Google Scholar]
- 20.West J, Rentzeperis F, Adam C, Bravo R, Luddy KA, Robertson-Tessi M, et al. Tumor-Immune Metaphenotypes Orchestrate an Evolutionary Bottleneck That Promotes Metabolic Transformation. Frontiers in Immunology 2024; 15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bekker RA, Zahid MU, Binning JM, Spring BQ, Hwu P, Pilon-Thomas S, et al. Rethinking the Immunotherapy Numbers Game. Journal for Immunotherapy of Cancer 2022; 10:e005107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Murias-Closas A, Prats C, Calvo G, López-Codina D, and Olesti E. Computational Modelling of CAR T-cell Therapy: From Cellular Kinetics to Patient-Level Predictions. eBioMedicine 2025; 113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gatenby RA and Brown JS. Integrating Evolutionary Dynamics into Cancer Therapy. Nature Reviews Clinical Oncology 2020; 17:675–86 [DOI] [PubMed] [Google Scholar]
- 24.Corral-Acero J, Margara F, Marciniak M, Rodero C, Loncaric F, Feng Y, et al. The ‘Digital Twin’ to Enable the Vision of Precision Cardiology. European Heart Journal 2020; 41:4556–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Brady R and Enderling H. Mathematical Models of Cancer: When to Predict Novel Therapies, and When Not To. Bulletin of Mathematical Biology 2019; 81:3722–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kim E, Rebecca VW, Smalley KS, and Anderson ARA. Phase i trials in melanoma: A framework to translate preclinical findings to the clinic. European Journal of Cancer 2016; 67:213–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Norton L and Simon R. Tumor Size, Sensitivity to Therapy, and Design of Treatment Schedules. Cancer Treatment Reports 1977; 61:1307–17 [PubMed] [Google Scholar]
- 28.McMahon SJ. The linear quadratic model: usage, interpretation and challenges. Physics in Medicine & Biology 2018; 64:01TR01. [DOI] [PubMed] [Google Scholar]
- 29.Stein AM, Grupp SA, Levine JE, Laetsch TW, Pulsipher MA, Boyer MW, et al. Tisagenlecleucel Model-Based Cellular Kinetic Analysis of Chimeric Antigen Receptor-T Cells. CPT: Pharmacometrics & Systems Pharmacology 2019; 8:285–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Skipper HE, Schabel FM Jr, and Wilcox WS. Experimental Evaluation of Potential Anticancer Agents. XIII. On the Criteria and Kinetics Associated with “Curability” of Experimental Leukemia. Cancer Chemotherapy Reports 1964; 35:1–111 [PubMed] [Google Scholar]
- 31.Benzekry S Artificial Intelligence and Mechanistic Modeling for Clinical Decision Making in Oncology. Clinical Pharmacology & Therapeutics 2020; 108:471–86 [DOI] [PubMed] [Google Scholar]
- 32.McKenna MT, Weis JA, Brock A, Quaranta V, and Yankeelov TE. Precision Medicine with Imprecise Therapy: Computational Modeling for Chemotherapy in Breast Cancer. Translational Oncology 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.The Chemotherapy Source Book. 4th ed. Philadelphia: Lippincott Williams & Wilkins, 2008 [Google Scholar]
- 34.Frei E and Freireich EJ. Progress and Perspectives in the Chemotherapy of Acute Leukemia. Advances in Chemotherapy. 1965; 2:269–98 [DOI] [PubMed] [Google Scholar]
- 35.Pomeroy AE, Schmidt EV, Sorger PK, and Palmer AC. Drug independence and the curability of cancer by combination chemotherapy. Trends in Cancer 2022; 8:915–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Freireich EJ, Karon M, Frei E III, et al. Quadruple combination therapy (VAMP) for acute lymphocytic leukemia of childhood. Proc Am Assoc Cancer Res Vol. 5. 20. 1964. :8 [Google Scholar]
- 37.Pui CH and Evans WE. A 50-Year Journey to Cure Childhood Acute Lymphoblastic Leukemia. Seminars in Hematology. 2013; 50:185–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Michor F and Beal K. Improving Cancer Treatment via Mathematical Modeling: Surmounting the Challenges Is Worth the Effort. Cell 2015; 163:1059–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Norton L Theoretical Concepts and the Emerging Role of Taxanes in Adjuvant Therapy. The Oncologist 2001; 6:30–5 [DOI] [PubMed] [Google Scholar]
- 40.Citron ML, Berry DA, Cirrincione C, Hudis C, Winer EP, Gradishar WJ, et al. Randomized Trial of Dose-Dense Versus Conventionally Scheduled and Sequential Versus Concurrent Combination Chemotherapy as Postoperative Adjuvant Treatment of Node-Positive Primary Breast Cancer: First Report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741. Journal of Clinical Oncology 2003; 21:1431–9 [DOI] [PubMed] [Google Scholar]
- 41.Burstein HJ. Selection and administration of adjuvant chemotherapy for HER2-negative breast cancer. UpToDate. Waltham, MA: Wolters Kluwer, 2025 [Google Scholar]
- 42.Traina TA, Dugan U, Higgins B, Kolinsky K, Theodoulou M, Hudis CA, et al. Optimizing Chemotherapy Dose and Schedule by Norton-Simon Mathematical Modeling. Breast Disease 2010; 31:7–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Traina TA, Theodoulou M, Feigin K, Patil S, Tan KL, Edwards C, et al. Phase I Study of a Novel Capecitabine Schedule Based on the Norton-Simon Mathematical Model in Patients With Metastatic Breast Cancer. Journal of Clinical Oncology 2008; 26:1797–802 [DOI] [PubMed] [Google Scholar]
- 44.Gajria D, Feigin K, Tan LK, Patil S, Geneus S, Theodoulou M, et al. Phase 2 trial of a novel capecitabine dosing schedule in combination with bevacizumab for patients with metastatic breast cancer. Cancer 2011; 117:4125–31 [DOI] [PubMed] [Google Scholar]
- 45.Gajria D, Gonzalez J, Feigin K, Patil S, Chen C, Theodoulou M, et al. Phase II trial of a novel capecitabine dosing schedule in combination with lapatinib for the treatment of patients with HER2-positive metastatic breast cancer. Breast Cancer Research and Treatment 2012; 131:111–6 [DOI] [PubMed] [Google Scholar]
- 46.Morikawa A, de Stanchina E, Pentsova E, Kemeny MM, Li BT, Tang K, et al. Phase I Study of Intermittent High-Dose Lapatinib Alternating with Capecitabine for HER2-Positive Breast Cancer Patients with Central Nervous System Metastases. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research 2019; 25:3784–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Khan QJ, Bohnenkamp C, Monson TE, Phadnis MA, Clark L, Smith HE, et al. Randomized Trial of Fixed-Dose Capecitabine Compared With Standard-Dose Capecitabine in Metastatic Breast Cancer: X-7/7 Trial. JCO Oncology Advances 2025. :e2400068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Joiner MC and Kogel A van der. Basic Clinical Radiobiology. 4th ed. London: CRC Press, 2009 [Google Scholar]
- 49.van Leeuwen CM, Oei AL, Crezee J, Bel A, Franken NAP, Stalpers LJA, et al. The Alfa and Beta of Tumours: A Review of Parameters of the Linear-Quadratic Model, Derived from Clinical Radiotherapy Studies. Radiation Oncology 2018; 13:96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Douglas BG and Fowler JF. The Effect of Multiple Small Doses of x Rays on Skin Reactions in the Mouse and a Basic Interpretation. Radiation Research 1976; 66:401–26 [PubMed] [Google Scholar]
- 51.Williams MV, Denekamp J, and Fowler JF. A Review of α β Ratios for Experimental Tumors: Implications for Clinical Studies of Altered Fractionation. International Journal of Radiation Oncology, Biology, Physics 1985; 11:87–96 [DOI] [PubMed] [Google Scholar]
- 52.Bourhis J, Overgaard J, Audry H, Ang KK, Saunders M, Bernier J, et al. Hyperfractionated or Accelerated Radiotherapy in Head and Neck Cancer: A Meta-Analysis. The Lancet 2006; 368:843–54 [DOI] [PubMed] [Google Scholar]
- 53.Leder K, Pitter K, LaPlant Q, Hambardzumyan D, Ross BD, Chan TA, et al. Mathematical Modeling of PDGF-Driven Glioblastoma Reveals Optimized Radiation Dosing Schedules. Cell 2014; 156:603–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dean JA, Tanguturi SK, Cagney D, Shin KY, Youssef G, Aizer A, et al. Phase I study of a novel glioblastoma radiation therapy schedule exploiting cell-state plasticity. Neuro-Oncology 2023; 25:1100–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.López Alfonso JC, Parsai S, Joshi N, Godley A, Shah C, Koyfman SA, et al. Temporally feathered intensity-modulated radiation therapy: A planning technique to reduce normal tissue toxicity. Medical Physics 2018; 45:3466–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Parsai S, Qiu RLJ, Qi P, Sedor G, Fuller CD, Murray E, et al. In vivo assessment of the safety of standard fractionation Temporally Feathered Radiation Therapy (TFRT) for head and neck squamous cell carcinoma: An R-IDEAL Stage 1/2a first-in-humans/feasibility demonstration of new technology implementation. Radiotherapy and Oncology 2021; 163:39–45 [DOI] [PubMed] [Google Scholar]
- 57.Scott JG, Sedor G, Ellsworth P, Scarborough JA, Ahmed KA, Oliver DE, et al. Pan-Cancer Prediction of Radiotherapy Benefit Using Genomic-Adjusted Radiation Dose (GARD): A Cohort-Based Pooled Analysis. The Lancet Oncology 2021; 22:1221–9 [DOI] [PubMed] [Google Scholar]
- 58.Torres-Roca JF, Eschrich S, Zhao H, Bloom G, Sung J, McCarthy S, et al. Prediction of Radiation Sensitivity Using a Gene Expression Classifier. Cancer Research 2005; 65:7169–76 [DOI] [PubMed] [Google Scholar]
- 59.Naghavi AO, Bryant JM, Kim Y, Weygand J, Redler G, Sim AJ, et al. Habitat Escalated Adaptive Therapy (HEAT): A Phase 2 Trial Utilizing Radiomic Habitat-Directed and Genomic-Adjusted Radiation Dose (GARD) Optimization for High-Grade Soft Tissue Sarcoma. BMC Cancer 2024; 24:437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Prokopiou S, Moros EG, Poleszczuk J, Caudell J, Torres-Roca JF, Latifi K, et al. A proliferation saturation index to predict radiation response and personalize radiotherapy fractionation. Radiation Oncology 2015; 10:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Caudell J Use of a Proliferation Saturation Index to Determine Personalized Radiotherapy Fractionation for Patients With HPV+ Oropharyngeal Cancers. 2021. Available from: https://cdn.clinicaltrials.gov/large-docs/33/NCT03656133/Prot_SAP_000.pdf [Accessed on: 2025 May 6] [Google Scholar]
- 62.Zahid MU, Yang GQ, Echevarria M, Caudell JJ, and Enderling H. Proliferation Saturation Index to Simulate Adaptive Radiation Fractionation in HPV-Associated Oropharyngeal Cancer. International Journal of Radiation Oncology, Biology, Physics. ASTRO 2023: 65th Annual Meeting 2023; 117:e495–e496 [Google Scholar]
- 63.Sorger PK, Allerheiligen SR, Abernethy DR, Altman RB, Brouwer KL, Califano A, et al. Quantitative and systems pharmacology in the post-genomic era: new approaches to discovering drugs and understanding therapeutic mechanisms. An NIH white paper by the QSP workshop group. Bethesda (MD): NIH. 2011:1–47 [Google Scholar]
- 64.Bai JP, Liu G, Zhao M, Wang J, Xiong Y, Truong T, et al. Landscape of regulatory quantitative systems pharmacology submissions to the US Food and Drug Administration: An update report. CPT: Pharmacometrics & Systems Pharmacology 2024; 13:2102–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Azer K, Kaddi CD, Barrett JS, Bai JPF, McQuade ST, Merrill NJ, et al. History and Future Perspectives on the Discipline of Quantitative Systems Pharmacology Modeling and Its Applications. Frontiers in Physiology. 2021; 12:637999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Garralda E, Dienstmann R, and Tabernero J. Pharmacokinetic/Pharmacodynamic Modeling for Drug Development in Oncology. American Society of Clinical Oncology Educational Book 2018; 37:210–5 [DOI] [PubMed] [Google Scholar]
- 67.Agema BC, Koch BCP, Mathijssen RHJ, and Koolen SLW. From Prospective Evaluation to Practice: Model-Informed Dose Optimization in Oncology. Drugs 2025; 85:487–503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Calvert AH, Newell DR, Gumbrell LA, O’Reilly S, Burnell M, Boxall FE, et al. Carboplatin dosage: prospective evaluation of a simple formula based on renal function. Journal of Clinical Oncology 1989; 7:1748–56 [DOI] [PubMed] [Google Scholar]
- 69.Evans WE, Relling MV, Rodman JH, Crom WR, Boyett JM, and Pui CH. Conventional Compared with Individualized Chemotherapy for Childhood Acute Lymphoblastic Leukemia. The New England Journal of Medicine 1998; 338:499–505 [DOI] [PubMed] [Google Scholar]
- 70.Bleyzac N, Souillet G, Magron P, Janoly A, Martin P, Bertrand Y, et al. Improved clinical outcome of paediatric bone marrow recipients using a test dose and Bayesian pharmacokinetic individualization of busulfan dosage regimens. Bone Marrow Transplantation 2001; 28:743–51 [DOI] [PubMed] [Google Scholar]
- 71.Jonge MEd, van den Bongard HJGD, Huitema ADR, Mathôt RAA, Rosing H, Baas P, et al. Bayesian Pharmacokinetically Guided Dosing of Paclitaxel in Patients with Non-Small Cell Lung Cancer. Clinical Cancer Research 2004; 10:2237–44 [DOI] [PubMed] [Google Scholar]
- 72.Yamamoto N, Tamura T, Murakami H, Shimoyama T, Nokihara H, Ueda Y, et al. Randomized Pharmacokinetic and Pharmacodynamic Study of Docetaxel: Dosing Based on Body-Surface Area Compared With Individualized Dosing Based on Cytochrome P450 Activity Estimated Using a Urinary Metabolite of Exogenous Cortisol. Journal of Clinical Oncology 2005; 23:1061–9 [DOI] [PubMed] [Google Scholar]
- 73.Wall A, Gajjar A, Link A, Mahmoud H, Pui CH, and Relling M. Individualized methotrexate dosing in children with relapsed acute lymphoblastic leukemia. Leukemia 2000; 14:221–5 [DOI] [PubMed] [Google Scholar]
- 74.Gamelin E, Delva R, Jacob J, Merrouche Y, Raoul JL, Pezet D, et al. Individual Fluorouracil Dose Adjustment Based on Pharmacokinetic Follow-Up Compared With Conventional Dosage: Results of a Multicenter Randomized Trial of Patients With Metastatic Colorectal Cancer. Journal of Clinical Oncology 2008; 26:2099–105 [DOI] [PubMed] [Google Scholar]
- 75.Salas S, Mercier C, Ciccolini J, Pourroy B, Fanciullino R, Tranchand B, et al. Therapeutic Drug Monitoring for Dose Individualization of Cisplatin in Testicular Cancer Patients Based Upon Total Platinum Measurement in Plasma. Therapeutic Drug Monitoring 2006; 28:532–9 [DOI] [PubMed] [Google Scholar]
- 76.Long-Boyle JR, Savic R, Yan S, Bartelink I, Musick L, French D, et al. Population Pharmacokinetics of Busulfan in Pediatric and Young Adult Patients Undergoing Hematopoietic Cell Transplant. Therapeutic Drug Monitoring 2015; 37:236–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hénin E, Meille C, Barbolosi D, You B, Guitton J, Iliadis A, et al. Revisiting dosing regimen using PK/PD modeling: the MODEL1 phase I/II trial of docetaxel plus epirubicin in metastatic breast cancer patients. Breast Cancer Research and Treatment 2016; 156:331–41 [DOI] [PubMed] [Google Scholar]
- 78.Meille C, Barbolosi D, Ciccolini J, Freyer G, and Iliadis A. Revisiting Dosing Regimen Using Pharmacokinetic/Pharmacodynamic Mathematical Modeling: Densification and Intensification of Combination Cancer Therapy. Clinical Pharmacokinetics 2016; 55:1015–25 [DOI] [PubMed] [Google Scholar]
- 79.Engels FK, Loos WJ, Bol JMvd, Bruijn Pd, Mathijssen RH, Verweij J, et al. Therapeutic Drug Monitoring for the Individualization of Docetaxel Dosing: A Randomized Pharmacokinetic Study. Clinical Cancer Research 2011; 17:353–62 [DOI] [PubMed] [Google Scholar]
- 80.Barlesi F, Imbs DC, Tomasini P, Greillier L, Galloux M, Testot-Ferry A, et al. Mathematical modeling for Phase I cancer trials: A study of metronomic vinorelbine for advanced non-small cell lung cancer (NSCLC) and mesothelioma patients. Oncotarget 2017; 8:47161–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Barlesi F, Deyme L, Imbs DC, Cousin E, Barbolosi M, Bonnet S, et al. Revisiting metronomic vinorelbine with mathematical modelling: a Phase I trial in lung cancer. Cancer Chemotherapy and Pharmacology 2022; 90:149–60 [DOI] [PubMed] [Google Scholar]
- 82.Goldie JH and Coldman AJ. A mathematic model for relating the drug sensitivity of tumors to their spontaneous mutation rate. Cancer treatment reports 1979; 63:1727–33 [PubMed] [Google Scholar]
- 83.Coldman AJ and Goldie JH. A stochastic model for the origin and treatment of tumors containing drug-resistant cells. Bulletin of Mathematical Biology 1986; 48:279–92 [DOI] [PubMed] [Google Scholar]
- 84.Murakami Y, Uemura K, Sudo T, Hashimoto Y, Kondo N, Nakagawa N, et al. Early initiation of adjuvant chemotherapy improves survival of patients with pancreatic carcinoma after surgical resection. Cancer Chemotherapy and Pharmacology 2013; 71:419–29 [DOI] [PubMed] [Google Scholar]
- 85.Gögenur M, Rosen AW, Iveson T, Kerr RS, Saunders MP, Cassidy J, et al. Time from colorectal cancer surgery to adjuvant chemotherapy: post hoc analysis of the SCOT randomized clinical trial. JAMA Surgery 2024; 159:865–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gagliato DdM, Gonzalez-Angulo AM, Lei X, Theriault RL, Giordano SH, Valero V, et al. Clinical Impact of Delaying Initiation of Adjuvant Chemotherapy in Patients With Breast Cancer. Journal of Clinical Oncology 2014; 32:735–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Abdel-Razeq H, Mansour A, Edaily S, and Dayyat A. Delays in Initiating Anti-Cancer Therapy for Early-Stage Breast Cancer—How Slow Can We Go? Journal of Clinical Medicine 2023; 12:4502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rhodin KE, Raman V, Jawitz OK, Tong BC, Harpole DH, and D’Amico TA. The effect of timing of adjuvant therapy on survival after esophagectomy. The Annals of Thoracic Surgery 2020; 110:1023–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Liu Y, Zhai X, Li J, Li Z, Ma D, and Wang Z. Is there an optimal time to initiate adjuvant chemotherapy to predict benefit of survival in non-small cell lung cancer? Chinese Journal of Cancer Research 2017; 29:263–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lampa E, Boros M, Berglund A, Wagenius G, and Oskarsdottir GN. Timing of Adjuvant Chemotherapy After Surgical Resection for Non-small Cell Lung Cancer: A Target Trial Emulation Using Nationwide Swedish Registry Data. Annals of Surgical Oncology 2025. :1–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Goldie J, Coldman A, and Gudauskas G. Rationale for the use of alternating non-cross-resistant chemotherapy. Cancer Treat Rep 1982; 66:439–49 [PubMed] [Google Scholar]
- 92.Bonadonna G Sequential or Alternating Doxorubicin and CMF Regimens in Breast Cancer With More Than Three Positive Nodes: Ten-Year Results. JAMA 1995; 273:542. [PubMed] [Google Scholar]
- 93.Paridaens R, Van Aelst F, Georgoulias V, Samonnig H, Cocquyt V, Zielinski C, et al. A randomized phase II study of alternating and sequential regimens of docetaxel and doxorubicin as first-line chemotherapy for metastatic breast cancer. Annals of Oncology 2003; 14:433–40 [DOI] [PubMed] [Google Scholar]
- 94.Sandford NL, Searle J, and Kerr J. Successive waves of apoptosis in the rat prostate after repeated withdrawal of testosterone stimulation. Pathology 1984; 16:406–10 [DOI] [PubMed] [Google Scholar]
- 95.Akakura K, Bruchovsky N, Goldenberg SL, Rennie PS, Buckley AR, and Sullivan LD. Effects of intermittent androgen suppression on androgen-dependent tumors. Apoptosis and serum prostatespecific antigen. Cancer 1993; 71:2782–90 [DOI] [PubMed] [Google Scholar]
- 96.Bruchovsky N, Klotz L, Crook J, Malone S, Ludgate C, Morris WJ, et al. Final results of the Canadian prospective phase II trial of intermittent androgen suppression for men in biochemical recurrence after radiotherapy for locally advanced prostate cancer: clinical parameters. Cancer 2006; 107:389–95 [DOI] [PubMed] [Google Scholar]
- 97.Bruchovsky N, Klotz L, Crook J, and Goldenberg SL. Locally advanced prostate cancer—biochemical results from a prospective phase II study of intermittent androgen suppression for men with evidence of prostate-specific antigen recurrence after radiotherapy. Cancer: Interdisciplinary International Journal of the American Cancer Society 2007; 109:858–67 [DOI] [PubMed] [Google Scholar]
- 98.Bruchovsky N, Klotz L, Crook J, Phillips N, Abersbach J, and Goldenberg SL. Quality of life, morbidity, and mortality results of a prospective phase II study of intermittent androgen suppression for men with evidence of prostate-specific antigen relapse after radiation therapy for locally advanced prostate cancer. Clinical Genitourinary Cancer 2008; 6:46–52 [DOI] [PubMed] [Google Scholar]
- 99.Jackson TL. A mathematical investigation of the multiple pathways to recurrent prostate cancer: comparison with experimental data. Neoplasia 2004; 6:697–704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ideta AM, Tanaka G, Takeuchi T, and Aihara K. A mathematical model of intermittent androgen suppression for prostate cancer. Journal of Nonlinear Science 2008; 18:593–614 [Google Scholar]
- 101.Hirata Y, Bruchovsky N, and Aihara K. Development of a mathematical model that predicts the outcome of hormone therapy for prostate cancer. Journal of Theoretical Biology 2010; 264:517–27 [DOI] [PubMed] [Google Scholar]
- 102.Hirata Y, Tanaka G, Bruchovsky N, and Aihara K. Mathematically modelling and controlling prostate cancer under intermittent hormone therapy. Asian Journal of Andrology 2012; 14:270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Tanaka G, Hirata Y, Goldenberg SL, Bruchovsky N, and Aihara K. Mathematical modelling of prostate cancer growth and its application to hormone therapy. Philosophical Transactions of the Royal Society A: Mathematical, Physical and Engineering Sciences 2010; 368:5029–44 [DOI] [PubMed] [Google Scholar]
- 104.Suzuki T, Bruchovsky N, and Aihara K. Piecewise affine systems modelling for optimizing hormone therapy of prostate cancer. Philosophical Transactions of the Royal Society A: Mathematical, Physical and Engineering Sciences 2010; 368:5045–59 [DOI] [PubMed] [Google Scholar]
- 105.Shimada T and Aihara K. A nonlinear model with competition between prostate tumor cells and its application to intermittent androgen suppression therapy of prostate cancer. Mathematical Biosciences 2008; 214:134–9 [DOI] [PubMed] [Google Scholar]
- 106.Gatenby RA, Silva AS, Gillies RJ, and Frieden BR. Adaptive Therapy. Cancer Research 2009; 69:4894–903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zhang J, Cunningham JJ, Brown JS, and Gatenby RA. Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer. Nature Communications 2017; 8:1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang J, Cunningham J, Brown J, and Gatenby R. Evolution-based mathematical models significantly prolong response to abiraterone in metastatic castrate-resistant prostate cancer and identify strategies to further improve outcomes. eLife 2022; 11:e76284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Brady-Nicholls R, Nagy JD, Gerke TA, Zhang T, Wang AZ, Zhang J, et al. Prostate-specific antigen dynamics predict individual responses to intermittent androgen deprivation. Nature Communications 2020; 11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kim E, Brown JS, Eroglu Z, and Anderson AR. Adaptive Therapy for Metastatic Melanoma: Predictions from Patient Calibrated Mathematical Models. Cancers 2021; 13:823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kim E, Gallaher J, Robertson-Tessi M, Smalley I, Gatenby R, Smalley K, et al. Abstract SY38–02: Personalized adaptive therapy for metastatic cancer: An integrated approach. Cancer Research 2019; 79:SY38–02–SY38–02 [Google Scholar]
- 112.Maley CC, Seyedi S, Saha D, and Anderson A. Abstract A017: Computational and mouse models of adaptive therapy with multiple drugs in breast cancer. Cancer Research 2024; 84:A017–A017 [Google Scholar]
- 113.Bruno R, Bottino D, De Alwis DP, Fojo AT, Guedj J, Liu C, et al. Progress and Opportunities to Advance Clinical Cancer Therapeutics Using Tumor Dynamic Models. Clinical Cancer Research 2020; 26:1787–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Liu LL, Li F, Pao W, and Michor F. Dose-Dependent Mutation Rates Determine Optimum Erlotinib Dosing Strategies for EGFR Mutant Non-Small Cell Lung Cancer Patients. PLOS ONE 2015; 10:e0141665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Poels KE, Schoenfeld AJ, Makhnin A, Tobi Y, Wang Y, Frisco-Cabanos H, et al. Identification of optimal dosing schedules of dacomitinib and osimertinib for a phase I/II trial in advanced EGFR-mutant non-small cell lung cancer. Nature Communications 2021; 12:3697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chmielecki J, Foo J, Oxnard GR, Hutchinson K, Ohashi K, Somwar R, et al. Optimization of dosing for EGFR-mutant non–small cell lung cancer with evolutionary cancer modeling. Science Translational Medicine 2011; 3:90ra59–90ra59 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Foo J and Michor F. Evolution of Resistance to Targeted Anti-Cancer Therapies during Continuous and Pulsed Administration Strategies. PLOS Computational Biology 2009; 5:1–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Foo J and Michor F. Evolution of resistance to anti-cancer therapy during general dosing schedules. Journal of Theoretical Biology 2010; 263:179–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Yu H, Sima C, Feldman D, Liu L, Vaitheesvaran B, Cross J, et al. Phase 1 study of twice weekly pulse dose and daily low-dose erlotinib as initial treatment for patients with EGFR-mutant lung cancers. Annals of Oncology 2017; 28:278–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brown K, Comisar C, Witjes H, Maringwa J, Greef R de, Vishwanathan K, et al. Population pharmacokinetics and exposure-response of osimertinib in patients with non-small cell lung cancer. British Journal of Clinical Pharmacology 2017; 83:1216–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Plana D, Palmer AC, and Sorger PK. Independent drug action in combination therapy: implications for precision oncology. Cancer Discovery 2022; 12:606–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Loewe S The Problem of Synergism and Antagonism of Combined Drugs. Arzneimittel-Forschung. 1953; 3:285–90 [PubMed] [Google Scholar]
- 123.Bliss CI. The Toxicity of Poisons Applied Jointly. Annals of Applied Biology. 1939; 26:585–615 [Google Scholar]
- 124.Bozic I and Nowak MA. Resisting resistance. Annual Review of Cancer Biology 2017; 1:203–21 [Google Scholar]
- 125.Palmer AC and Sorger PK. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell 2017; 171:1678–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hwangbo H, Patterson SC, Dai A, Plana D, and Palmer AC. Additivity predicts the efficacy of most approved combination therapies for advanced cancer. Nature Cancer 2023; 4:1693–704 [DOI] [PubMed] [Google Scholar]
- 127.Sové RJ, Jafarnejad M, Zhao C, Wang H, Ma H, and Popel AS. QSP-IO: A Quantitative Systems Pharmacology Toolbox for Mechanistic Multiscale Modeling for Immuno-Oncology Applications. CPT: Pharmacometrics & Systems Pharmacology. 2020; 9:484–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gatenby RA, Artzy-Randrup Y, Epstein T, Reed DR, and Brown JS. Eradicating Metastatic Cancer and the Eco-Evolutionary Dynamics of Anthropocene Extinctions. Cancer Research 2020; 80:613–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Reed DR, Metts J, Pressley M, Fridley BL, Hayashi M, Isakoff MS, et al. An evolutionary framework for treating pediatric sarcomas. Cancer 2020; 126:2577–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Metts J, Russell T, Reed D, and Trucco M. A Proposed Trial Design for the Treatment of Widely Metastatic Ewing Sarcoma Inspired by Evolutionary Dynamics. Cancers 2022; 14:736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Fischer A, Vázquez-García I, and Mustonen V. The value of monitoring to control evolving populations. Proceedings of the National Academy of Sciences 2015; 112:1007–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Robertson-Tessi M, Brown JS, Poole MI, Johnson M, Marusyk A, Gallaher JA, et al. Feasibility of an Evolutionary Tumor Board for Generating Novel Personalized Therapeutic Strategies. 2023 [Google Scholar]
- 133.Oliveira G and Wu CJ. Dynamics and Specificities of T Cells in Cancer Immunotherapy. Nature Reviews Cancer 2023; 23:295–316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Madabushi R, Seo P, Zhao L, Tegenge M, and Zhu H. Review: Role of Model-Informed Drug Development Approaches in the Lifecycle of Drug Development and Regulatory Decision-Making. Pharmaceutical Research 2022; 39:1669–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Morrissey KM, Marchand M, Patel H, Zhang R, Wu B, Phyllis Chan H, et al. Alternative Dosing Regimens for Atezolizumab: An Example of Model-Informed Drug Development in the Post-marketing Setting. Cancer Chemotherapy and Pharmacology 2019; 84:1257–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Bi Y, Liu J, Furmanski B, Zhao H, Yu J, Osgood C, et al. Model-Informed Drug Development Approach Supporting Approval of the 4-Week (Q4W) Dosing Schedule for Nivolumab (Opdivo) across Multiple Indications: A Regulatory Perspective. Annals of Oncology 2019; 30:644–51 [DOI] [PubMed] [Google Scholar]
- 137.U.S. Food & Drug Administration. KYMRIAH. 2025. Available from: https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/kymriah [Accessed on: 2025 Sep 12] [Google Scholar]
- 138.Aghamiri SS, Amin R, and Helikar T. Recent applications of quantitative systems pharmacology and machine learning models across diseases. Journal of Pharmacokinetics and Pharmacodynamics 2022; 49:19–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Walker R and Enderling H. From concept to clinic: Mathematically informed immunotherapy. Current Problems in Cancer 2016; 40. SI: Abscopal Effect in Cancer Therapy:68–83 [DOI] [PubMed] [Google Scholar]
- 140.Chelliah V, Lazarou G, Bhatnagar S, Gibbs JP, Nijsen M, Ray A, et al. Quantitative Systems Pharmacology Approaches for Immuno-Oncology: Adding Virtual Patients to the Development Paradigm. Clinical Pharmacology & Therapeutics 2021; 109:605–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Lorenzo G, Ahmed SR, Hormuth DA, Vaughn B, Kalpathy-Cramer J, Solorio L, et al. Patient-Specific, Mechanistic Models of Tumor Growth Incorporating Artificial Intelligence and Big Data. Annual Review of Biomedical Engineering 2024; 26:529–60 [DOI] [PubMed] [Google Scholar]
- 142.Metzcar J, Jutzeler CR, Macklin P, Köhn-Luque A, and Brüningk SC. A review of mechanistic learning in mathematical oncology. Frontiers in Immunology 2024; 15:1363144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Harkos C, Hadjigeorgiou AG, Voutouri C, Kumar AS, Stylianopoulos T, and Jain RK. Using Mathematical Modelling and AI to Improve Delivery and Efficacy of Therapies in Cancer. Nature Reviews Cancer 2025; 25:324–40 [DOI] [PubMed] [Google Scholar]
- 144.Wang H, Arulraj T, Ippolito A, and Popel AS. From Virtual Patients to Digital Twins in Immuno-Oncology: Lessons Learned from Mechanistic Quantitative Systems Pharmacology Modeling. npj Digital Medicine 2024; 7:1–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Craig M, Gevertz JL, Kareva I, and Wilkie KP. A Practical Guide for the Generation of Model-Based Virtual Clinical Trials. Frontiers in Systems Biology 2023; 3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Creemers JHA, Ankan A, Roes KCB, Schröder G, Mehra N, Figdor CG, et al. In Silico Cancer Immunotherapy Trials Uncover the Consequences of Therapy-Specific Response Patterns for Clinical Trial Design and Outcome. Nature Communications 2023; 14:2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wang H, Sové RJ, Jafarnejad M, Rahmeh S, Jaffee EM, Stearns V, et al. Conducting a Virtual Clinical Trial in HER2-Negative Breast Cancer Using a Quantitative Systems Pharmacology Model With an Epigenetic Modulator and Immune Checkpoint Inhibitors. Frontiers in Bioengineering and Biotechnology 2020; 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.De Domenico M, Allegri L, Caldarelli G, d’Andrea V, Di Camillo B, Rocha LM, et al. Challenges and Opportunities for Digital Twins in Precision Medicine from a Complex Systems Perspective. npj Digital Medicine 2025; 8:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Hernandez-Boussard T, Macklin P, Greenspan EJ, Gryshuk AL, Stahlberg E, Syeda-Mahmood T, et al. Digital twins for predictive oncology will be a paradigm shift for precision cancer care. Nature Medicine 2021; 27:2065–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Laubenbacher R, Mehrad B, Shmulevich I, and Trayanova N. Digital twins in medicine. Nature Computational Science 2024; 4:184–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Stahlberg EA, Abdel-Rahman M, Aguilar B, Asadpoure A, Beckman RA, Borkon LL, et al. Exploring approaches for predictive cancer patient digital twins: Opportunities for collaboration and innovation. Frontiers in Digital Health 2022; 4:1007784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Swanson KR, Bridge C, Murray JD, and Alvord EC. Virtual and Real Brain Tumors: Using Mathematical Modeling to Quantify Glioma Growth and Invasion. Journal of the Neurological Sciences 2003; 216:1–10 [DOI] [PubMed] [Google Scholar]
- 153.Swanson KR, Rostomily RC, and Alvord EC. A Mathematical Modelling Tool for Predicting Survival of Individual Patients Following Resection of Glioblastoma: A Proof of Principle. British Journal of Cancer 2008; 98:113–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Rockne R, Rockhill JK, Mrugala M, Spence AM, Kalet I, Hendrickson K, et al. Predicting the Efficacy of Radiotherapy in Individual Glioblastoma Patients in Vivo: A Mathematical Modeling Approach. Physics in Medicine and Biology 2010; 55:3271–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Singhal A, Zhao X, Wall P, So E, Calderini G, Partin A, et al. The Hallmarks of Predictive Oncology. Cancer Discovery 2025; 15:271–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Elemento O, Khozin S, and Sternberg CN. The Use of Artificial Intelligence for Cancer Therapeutic Decision-Making. NEJM AI 2025; 2:AIra2401164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Benzekry S, Sentis C, Coze C, Tessonnier L, and André N. Development and Validation of a Prediction Model of Overall Survival in High-Risk Neuroblastoma Using Mechanistic Modeling of Metastasis. JCO Clinical Cancer Informatics. 2021; 5:81–90 [DOI] [PubMed] [Google Scholar]
- 158.Scibilia KR, Schlicke P, Schneller F, and Kuttler C. Predicting Resistance and Pseudoprogression: Are Minimalistic Immunoediting Mathematical Models Capable of Forecasting Checkpoint Inhibitor Treatment Outcomes in Lung Cancer? Mathematical Biosciences 2024; 376:109287. [DOI] [PubMed] [Google Scholar]
- 159.Ezhov I, Scibilia K, Franitza K, Steinbauer F, Shit S, Zimmer L, et al. Learn-Morph-Infer: A New Way of Solving the Inverse Problem for Brain Tumor Modeling. Medical Image Analysis 2023; 83:102672. [DOI] [PubMed] [Google Scholar]
- 160.Balcerak M, Weidner J, Karnakov P, Ezhov I, Litvinov S, Koumoutsakos P, et al. Individualizing Glioma Radiotherapy Planning by Optimization of a Data and Physics-Informed Discrete Loss. Nature Communications. 2025; 16:5982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Brunton SL, Proctor JL, and Kutz JN. Discovering Governing Equations from Data by Sparse Identification of Nonlinear Dynamical Systems. Proceedings of the National Academy of Sciences of the United States of America. 2016; 113:3932–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Brummer AB, Xella A, Woodall R, Adhikarla V, Cho H, Gutova M, et al. Data Driven Model Discovery and Interpretation for CAR T-cell Killing Using Sparse Identification and Latent Variables. Frontiers in Immunology. 2023; 14:1115536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Raissi M, Perdikaris P, and Karniadakis GE. Physics-Informed Neural Networks: A Deep Learning Framework for Solving Forward and Inverse Problems Involving Nonlinear Partial Differential Equations. Journal of Computational Physics 2019; 378:686–707 [Google Scholar]
- 164.Gallagher K, Strobl MA, Park DS, Spoendlin FC, Gatenby RA, Maini PK, et al. Mathematical Model-Driven Deep Learning Enables Personalized Adaptive Therapy. Cancer Research 2024; 84:1929–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Weaver DT, King ES, Maltas J, and Scott JG. Reinforcement learning informs optimal treatment strategies to limit antibiotic resistance. Proceedings of the National Academy of Sciences 2024; 121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Stein AM and Looby M. Benchmarking QSP Models Against Simple Models: A Path to Improved Comprehension and Predictive Performance. CPT: pharmacometrics & systems pharmacology. 2018; 7:487–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ross JS, Lehman R, and Gross CP. The Importance of Clinical Trial Data Sharing: Toward More Open Science. Circulation Cardiovascular Quality and Outcomes. 2012; 5:238–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Boeckhout M, Zielhuis GA, and Bredenoord AL. The FAIR guiding principles for data stewardship: fair enough? European Journal of Human Genetics 2018; 26:931–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Martorana M, Kuhn T, Siebes R, and van Ossenbruggen J. Aligning restricted access data with FAIR: a systematic review. PeerJ Computer Science 2022; 8:e1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Hallinan CM, Ward R, Hart GK, Sullivan C, Pratt N, Ng AP, et al. Seamless EMR Data Access: Integrated Governance, Digital Health and the OMOP-CDM. BMJ health & care informatics. 2024; 31:e100953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Yankeelov TE, Hormuth DA, Lima EA, Lorenzo G, Wu C, Okereke LC, et al. Designing clinical trials for patients who are not average. iScience 2024; 27:108589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.European Medicines Agency. Guideline on assessment and reporting of mechanistic models used in the context of model informed drug development. 2025. Available from: https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/yescarta [Accessed on: 2025 Apr 9] [Google Scholar]
- 173.U.S. Food & Drug Administration - Oncology Center of Excellence. Project Optimus - Reforming the Dose Optimization and Dose Selection Paradigm in Oncology. 2024. Available from: https://www.fda.gov/about-fda/oncology-center-excellence/project-optimus [Accessed on: 2025 Apr 9] [Google Scholar]
- 174.U.S. Food & Drug Administration. Drug Development Tools: Fit-for-Purpose Initiative. 2025. Available from: https://www.fda.gov/drugs/development-approval-process-drugs/drugdevelopment-tools-fit-purpose-initiative [Accessed on: 2025 Apr 9] [Google Scholar]
- 175.U.S. Food & Drug Administration. FDA Announces Plan to Phase Out Animal Testing Requirement for Monoclonal Antibodies and Other Drugs. visited on 2025-08-28. 2025. Available from: https://www.fda.gov/news-events/press-announcements/fda-announces-plan-phaseout-animal-testing-requirement-monoclonal-antibodies-and-other-drugs [Google Scholar]
- 176.Mak IW, Evaniew N, and Ghert M. Lost in Translation: Animal Models and Clinical Trials in Cancer Treatment. American Journal of Translational Research. 2014; 6:114–8 [PMC free article] [PubMed] [Google Scholar]
- 177.Nguyen DT, Famiglietti JE, Smolchek RA, Dupee Z, Diodati N, Pedro DI, et al. 3D In Vitro Platform for Cell and Explant Culture in Liquid-like Solids. Cells. 2022; 11:967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Pallmann P, Bedding AW, Choodari-Oskooei B, Dimairo M, Flight L, Hampson LV, et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Medicine 2018; 16:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Berry DA. Bayesian Clinical Trials. Nature Reviews Drug Discovery 2006; 5:27–36 [DOI] [PubMed] [Google Scholar]
- 180.Thall PF. Bayesian Utility-Based Designs for Subgroup-Specific Treatment Comparison and Early-Phase Dose Optimization in Oncology Clinical Trials. JCO Precision Oncology 2019. :1–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Wang H and Yee D. I-SPY 2: A Neoadjuvant Adaptive Clinical Trial Designed to Improve Outcomes in High-Risk Breast Cancer. Current Breast Cancer Reports. 2019; 11:303–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, et al. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women With Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncology 2020; 6:676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Anderson ARA. IMO Workshop - A History. 2020. Available from: https://mathematicaloncology.org/blog/IMO-workshop-a-history.html [Accessed on: 2025 Apr 9] [Google Scholar]
