Next Article in Journal
Solving Boundary Value Problems for a Class of Differential Equations Based on Elastic Transformation and Similar Construction Methods
Previous Article in Journal
AI-Assisted Game Theory Approaches to Bid Pricing Under Uncertainty in Construction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mathematical Modelling of Cancer Treatments, Resistance, Optimization

Department of Mechanical Engineering, Florida State University, Tallahassee, FL 32310, USA
AppliedMath 2025, 5(2), 40; https://doi.org/10.3390/appliedmath5020040
Submission received: 11 December 2024 / Revised: 16 February 2025 / Accepted: 21 February 2025 / Published: 4 April 2025

Abstract

:
Mathematical modeling plays a crucial role in the advancement of cancer treatments, offering a sophisticated framework for analyzing and optimizing therapeutic strategies. This approach employs mathematical and computational techniques to simulate diverse aspects of cancer therapy, including the effectiveness of various treatment modalities such as chemotherapy, radiation therapy, targeted therapy, and immunotherapy. By incorporating factors such as drug pharmacokinetics, tumor biology, and patient-specific characteristics, these models facilitate predictions of treatment responses and outcomes. Furthermore, mathematical models elucidate the mechanisms behind cancer treatment resistance, including genetic mutations and microenvironmental changes, thereby guiding researchers in designing strategies to mitigate or overcome resistance. The application of optimization techniques allows for the development of personalized treatment regimens that maximize therapeutic efficacy while minimizing adverse effects, taking into account patient-related variables such as tumor size and genetic profiles. This study elaborates on the key applications of mathematical modeling in oncology, encompassing the simulation of various cancer treatment modalities, the elucidation of resistance mechanisms, and the optimization of personalized treatment regimens. By integrating mathematical insights with experimental data and clinical observations, mathematical modeling emerges as a powerful tool in oncology, contributing to the development of more effective and personalized cancer therapies that improve patient outcomes.

1. Introduction

Mathematical modeling of cancer treatments (see Figure 1), resistance, and optimization involves using mathematical and computational techniques to simulate and analyze various aspects of cancer therapy. This approach helps researchers and clinicians understand how different treatment strategies affect tumor growth, how tumors develop resistance to therapy, and how to optimize treatment regimens to improve patient outcomes [1,2,3,4].
Mathematical models can simulate the effects of different treatment modalities such as chemotherapy, radiation therapy, targeted therapy, immunotherapy, and combination therapies. These models take into account factors such as drug pharmacokinetics and pharmacodynamics, tumor biology, and patient-specific characteristics to predict treatment responses and outcomes [5,6,7,8].
Cancer cells can develop resistance to treatment over time, leading to treatment failure and disease progression [9,10,11,12]. Mathematical models can elucidate the underlying mechanisms of resistance, such as genetic mutations, clonal selection, and microenvironmental changes. By understanding these mechanisms, researchers can identify strategies to overcome or prevent resistance and improve treatment efficacy [13,14,15].
Mathematical optimization techniques can be applied to design personalized treatment regimens that maximize therapeutic efficacy while minimizing toxicity and side effects. These optimization models consider factors such as drug dosing schedules, treatment sequencing, drug combinations, and patient-specific variables (e.g., tumor size, genetic profile, treatment history) to tailor therapy to individual patients [16,17,18,19]. Mathematical modeling provides a powerful tool for cancer researchers and clinicians to explore the complex dynamics of cancer treatments, resistance, and optimization. By integrating mathematical insights with experimental data and clinical observations, mathematical oncology contributes to the development of more effective and personalized cancer therapies [20,21].
Through this study, we aim to underscore the transformative potential of mathematical modeling in oncology, illustrating how it not only aids in our understanding of cancer but also paves the way for more effective and individualized treatment strategies that hold the promise of improving patient outcomes in a field that is continually evolving (see Figure 2).
Specifically, the modeling approach used in this study combines several mathematical techniques to simulate and predict cancer treatment outcomes. A fundamental cancer growth model is built using a differential equation that captures the natural growth dynamics of cancer cells, accounting for factors like growth rate, maximum population size, and the level of resistance to treatment. This model is then extended to incorporate the impact of drug dosage, where the drug’s effect reduces the cancer cell population based on the dosage level and how effective the drug is. To predict treatment response, a logistic regression model is used, which estimates the probability of successful treatment based on patient-specific characteristics like age, biomarker levels, and tumor size. Finally, an optimization process is employed to find the best drug dosage by mathematically determining the dosage that results in the fewest cancer cells at the end of the treatment period. These methods together provide a comprehensive understanding of cancer treatment, from basic growth simulation to predicting outcomes and optimizing dosage.

1.1. Mathematical Modelling of Cancer Treatments

Mathematical modeling of cancer treatments involves using mathematical and computational methods to simulate and analyze how different treatment strategies affect tumor growth and response. This approach allows researchers and clinicians to explore the dynamics of cancer treatment regimens in a controlled and quantitative manner.
Mathematical models are essential tools in cancer treatment, effectively capturing the pharmacokinetics and pharmacodynamics of cancer drugs. By integrating parameters such as drug concentrations in blood plasma, uptake by tumor cells, and the effects on cell proliferation and death, these models can predict how drugs distribute and perform throughout the body. They also simulate the growth dynamics of tumors, considering factors like cell proliferation, death, and migration, and allowing researchers to anticipate tumor responses to various treatment modalities, such as chemotherapy and immunotherapy. This predictive capability enables the identification of optimal treatment strategies aimed at inhibiting tumor progression [22].
In addition to predicting drug effectiveness, mathematical optimization techniques are employed to design personalized treatment regimens tailored to individual patients. These optimization models account for factors such as drug dosing schedules, treatment sequencing, and combinations of drugs, striving to maximize therapeutic efficacy while minimizing side effects and toxicity. By integrating patient-specific data, including tumor characteristics and genetic profiles, mathematical models can guide clinicians in predicting treatment responses and determining the most effective therapies for each individual. Overall, the synergy of mathematical modeling with experimental data and clinical observations enhances the development of effective, personalized cancer treatment strategies, leading to improved outcomes and survival rates for patients [23].

1.2. Mathematical Formulation of Cancer Treatments

Mathematical modeling of cancer treatments involves using mathematical equations to represent the dynamics of tumor growth and response to various treatment modalities [24,25,26]. Here are some key aspects of mathematical modeling in cancer treatment, along with corresponding equations:

1.3. Tumor Growth Dynamics

The growth of a tumor can be described by mathematical models such as the Gompertz model or the logistic growth model [27,28]. The Gompertz model, for example, describes tumor growth as an exponential decrease in growth rate over time:
d V d t = r V × ln K V
where V represents the tumor volume, r is the growth rate constant, and K is the carrying capacity of the tumor.

1.4. Drug Pharmacokinetics and Pharmacodynamics

Pharmacokinetic models describe the concentration of drugs in the body over time, typically using compartmental models such as the one-compartment model or the two-compartment model [29]. For example, the one-compartment model can be represented by the equation:
d C d t = k × C
where C is the drug concentration in the central compartment, and k is the elimination rate constant. Pharmacodynamic models describe the effect of drugs on tumor cells, often using equations to represent drug–cell interactions and their impact on cell proliferation and death.
For instance, the Hill equation is commonly used to describe the dose–response relationship of drugs:
E = E m a x × C n E C 50 n + C n
where E is the drug effect, E m a x is the maximum effect, E C 50 n is the concentration at half-maximal effect, C is the drug concentration, and n is the Hill coefficient.

1.5. Optimization of Treatment Regimens

Optimization models aim to identify optimal treatment regimens by optimizing objective functions that balance treatment efficacy and toxicity. Linear programming and dynamic programming techniques can be used to optimize drug dosing schedules, treatment combinations, and sequencing strategies. For example, linear programming can be applied to maximize the therapeutic effect of combination therapy subject to constraints on toxicity and drug availability [30,31].

1.6. Personalized Medicine

Personalized medicine models incorporate patient-specific data, such as tumor characteristics and genetic profiles, to customize treatment decisions. Patient-specific parameters can be integrated into mathematical models to predict individual treatment responses and optimize personalized treatment plans. Mathematical modeling provides a quantitative framework for understanding and optimizing cancer treatments, allowing researchers and clinicians to design more effective and personalized therapeutic strategies [32,33,34].

2. Mathematical Modelling of Cancer Resistance

Mathematical modeling of cancer resistance involves studying the mechanisms by which tumors develop resistance to anticancer treatments, such as chemotherapy, targeted therapy, and immunotherapy. Here is an overview of how mathematical modeling is used to understand cancer resistance [35,36,37].

2.1. Evolutionary Dynamics

Cancer cells can evolve and adapt to selective pressures imposed by treatment, leading to the emergence of resistant subpopulations. Mathematical models, such as evolutionary game theory and population genetics models, are used to simulate the dynamics of tumor evolution and the emergence of resistant clones. These models incorporate factors such as mutation rates, fitness advantages conferred by resistance mutations, and competition between sensitive and resistant cell populations [38,39,40,41].

2.2. Drug Resistance Mechanisms

Mathematical models are used to investigate specific mechanisms of drug resistance, such as alterations in drug targets, activation of alternative signaling pathways, and changes in drug efflux mechanisms. Systems biology approaches, such as ordinary differential equations (ODEs) or agent-based models (ABMs), can be used to represent intracellular signaling networks and elucidate how perturbations in these networks lead to resistance [41,42].

2.3. Spatial Heterogeneity

Tumors are spatially heterogeneous environments, which can impact the development and spread of resistance. Spatial models, such as cellular automata or partial differential equations (PDEs), are employed to simulate tumor growth and treatment response in spatially structured environments. These models consider factors such as nutrient gradients, cell–cell interactions, and spatial distribution of treatment agents to predict how spatial heterogeneity influences the evolution of resistance [43,44].

2.4. Treatment Strategies to Overcome Resistance

Mathematical modeling is used to design treatment strategies aimed at overcoming or delaying the onset of resistance. Optimization models can identify optimal dosing schedules, drug combinations, and treatment sequences to minimize the likelihood of resistance emergence. Adaptive therapy approaches, which dynamically adjust treatment regimens based on tumor response, are investigated using mathematical models to exploit evolutionary principles and maintain treatment efficacy over time [45].

2.5. Clinical Translation

Mathematical models can inform clinical decision making by predicting patient-specific outcomes and guiding treatment selection. Patient-specific data, such as tumor genomics and imaging characteristics, are integrated into mathematical models to personalize treatment strategies and optimize therapeutic outcomes. Model-based predictions can help clinicians anticipate the likelihood of resistance development and adjust treatment plans accordingly to improve patient outcomes [46,47].
In general, mathematical modeling provides a powerful tool for studying cancer resistance mechanisms and designing effective strategies to overcome treatment resistance. By capturing the complex dynamics of tumor evolution and treatment response, mathematical models contribute to the development of more durable and personalized cancer therapies.

2.6. Mathematical Formulation of Cancer Resistance

Mathematical modeling of cancer resistance involves using mathematical equations to describe the dynamics of tumor growth, treatment response, and the emergence of resistant cell populations [48,49]. Here is how mathematical modeling is applied to study cancer resistance, along with examples of equations used in this context:

2.6.1. Population Dynamics Models

The logistic growth model describes tumor growth over time, considering factors such as proliferation rate and carrying capacity.
d N d t = r N ( 1 N K )
where N is the tumor cell population size, r is the growth rate, and K is the carrying capacity.
The Lotka–Volterra competition model models the competition between sensitive and resistant cell populations, incorporating growth and death rates for both populations.
d N 1 d t = r 1 N 1 ( 1 N 1 + α N 2 K 1 )
d N 2 d t = r 2 N 2 ( 1 N 2 + α N 1 K 2 )
where N 1 and N 2 are the sizes of the sensitive and resistant cell populations, respectively; r 1 and r 2 are the growth rates of the sensitive and resistant populations; α and β are the competitive effects of each population on the other; and K 1 and K 2 are the carrying capacities of the sensitive and resistant populations, respectively.

2.6.2. Pharmacokinetic-Pharmacodynamic (PK-PD) Models

The drug concentration model describes the concentration of a drug over time in the tumor microenvironment or systemic circulation.
d C d t = k e l i m . C
where C is the drug concentration, and k e l i m is the elimination rate constant.
The drug effect model describes the effect of a drug on tumor cells, considering factors such as drug potency and resistance mechanisms.
d E d t = k i n . C . E m a x k o u t . E
where E is the drug effect; k i n and k o u t are rate constants for drug uptake and elimination, respectively; and E m a x is the maximum drug effect achievable.

2.6.3. Evolutionary Models

The mutation-accumulation model describes the accumulation of mutations in cancer cells over time, leading to the emergence of resistant clones.
d N d t = μ N
where N is the number of cancer cells, and μ is the mutation rate.
The selection model models the growth advantage conferred by resistance mutations, leading to clonal expansion of resistant cells.
d N r e s i s t a n t d t = r r e s i s t a n t N r e s i s t a n t
where N r e s i s t a n t is the number of resistant cancer cells, and r r e s i s t a n t is the growth rate of resistant cells.
These equations, among others, are used in mathematical oncology to quantify the dynamics of cancer resistance and predict the effectiveness of different treatment strategies. By integrating experimental data and clinical observations into mathematical models, researchers can gain insights into the underlying mechanisms of cancer resistance and identify optimal treatment approaches to combat it.

3. Mathematical Optimization of Cancer Treatments and Resistance

Mathematical optimization techniques can be applied to optimize cancer treatments and address resistance [50,51]. Here is how mathematical optimization can be used in the context of cancer treatments and resistance:

3.1. Optimal Treatment Regimens

Optimal treatment regimens in cancer therapy can be determined through mathematical modeling and optimization techniques [52]. Here is how mathematical optimization can be used to identify the most effective treatment regimens:
Objective Function: Define an objective function that quantifies treatment efficacy and/or minimizes adverse effects. This function could incorporate variables such as tumor size reduction, patient survival rates, and toxicity levels.
Decision Variables: Identify decision variables that represent treatment parameters, such as drug dosages, administration schedules, and drug combinations. These variables can be continuous or discrete, depending on the treatment options available.
Constraints: Define constraints that reflect clinical considerations and limitations. These constraints may include maximum permissible drug doses, treatment duration, and physiological constraints based on patient characteristics.
Mathematical Model: Formulate a mathematical model that describes the dynamics of tumor growth, drug pharmacokinetics, and treatment response. This model can be based on differential equations, agent-based models, or other mathematical frameworks depending on the complexity of the system.
Optimization Algorithm: Apply optimization algorithms to solve the formulated optimization problem and identify the treatment regimen that maximizes the objective function while satisfying all constraints. Common optimization techniques include linear programming, nonlinear programming, genetic algorithms, and simulated annealing.
Sensitivity Analysis: Perform sensitivity analysis to assess the robustness of the optimal solution to variations in model parameters and assumptions. This analysis helps identify critical factors that influence treatment outcomes and guide decision making under uncertainty.
Clinical Validation: Validate the optimized treatment regimens through preclinical experiments, clinical trials, or retrospective analyses of patient data. This validation process ensures that the proposed regimens are clinically feasible and effective in real-world settings.
By mathematically optimizing treatment regimens, researchers and clinicians can systematically explore a vast space of possible interventions, identify optimal strategies tailored to individual patients, and ultimately improve treatment outcomes in cancer therapy.
Dose Optimization: Use optimization algorithms to determine the optimal dosage and scheduling of chemotherapy, immunotherapy, or targeted therapy to maximize tumor cell death while minimizing toxicity to healthy tissues.
Combination Therapy: Identify the most effective combinations of drugs and their respective dosages to overcome resistance mechanisms and enhance treatment efficacy.

3.2. Adaptive Therapy

Dynamic treatment strategies develop adaptive treatment protocols that adjust dosages and drug combinations in real time based on patient response and evolving tumor dynamics.
Optimal Control Theory: Apply optimal control theory to design treatment schedules that dynamically adapt to changes in tumor growth rates and drug sensitivity profiles.

3.3. Drug Development

Pharmacokinetic Optimization: Optimize drug properties such as bioavailability, distribution, and metabolism to enhance drug delivery to tumor sites and improve treatment efficacy. Pharmacokinetic mathematical optimization is a process used to optimize drug dosing regimens based on pharmacokinetic principles. Pharmacokinetics refers to the study of how drugs are absorbed, distributed, metabolized, and eliminated by the body over time. Mathematical optimization techniques are applied to determine the optimal drug doses and dosing schedules that achieve desired therapeutic outcomes while minimizing adverse effects [53,54].
Modeling Pharmacokinetics: Mathematical models are developed to describe the concentration–time profiles of drugs in the body. These models may involve differential equations that account for processes such as absorption, distribution, metabolism, and excretion of the drug.
Defining Objectives: The optimization process begins by defining specific therapeutic objectives, such as achieving target drug concentrations at the site of action or maintaining drug levels within a therapeutic window to maximize efficacy and minimize toxicity.
Identifying Parameters: Key pharmacokinetic parameters, such as drug clearance, volume of distribution, and absorption rate constants, are identified or estimated from experimental data or literature sources.
Formulating Optimization Problems: Optimization problems are formulated to find the optimal drug dosing regimens that best achieve the defined therapeutic objectives. These problems may involve maximizing drug efficacy, minimizing drug toxicity, or balancing trade-offs between multiple objectives.
Constraints: Constraints are imposed on the optimization problem to ensure that the proposed dosing regimens adhere to clinically relevant considerations, such as maximum permissible drug doses, dosing frequency, and physiological constraints based on patient characteristics.
Optimization Algorithms: Mathematical optimization algorithms, such as nonlinear programming, genetic algorithms, or gradient-based methods, are applied to solve the formulated optimization problems and identify the optimal drug dosing regimens.
Validation and Refinement: The optimized dosing regimens are validated through preclinical experiments, clinical trials, or retrospective analyses of patient data. The results are then used to refine the pharmacokinetic models and optimization strategies.
Pharmacokinetic mathematical optimization provides a systematic approach to design personalized dosing regimens that maximize drug efficacy, minimize toxicity, and improve therapeutic outcomes in clinical practice.
Objective Function: Maximize f(x)
where f(x) represents the objective function, which could be any relevant pharmacokinetic parameter or combination of parameters, such as maximum drug concentration, area under the curve, or time to reach steady-state concentration.
Constraints:
g i ( x ) b i h j ( x ) = c j
where g i ( x ) are inequality constraints that limit the feasible region of the optimization problem; h j ( x ) are equality constraints that must be satisfied; and b i and c j are constants representing the upper or lower bounds on the constraints. Variables x = ( x 1 , x 2 , , x n ) , where x represents the decision variables, which could include drug doses, dosing intervals, infusion rates, or other parameters related to drug administration.
The optimization problem can then be formulated as:
Maximize f(x)
Subject to:
g i ( x ) b i h j ( x ) = c j
Solving this optimization problem using appropriate mathematical optimization techniques will yield the optimal values of the decision variables x that maximize the objective function f(x) while satisfying the constraints. The specific form of the objective function and constraints will depend on the pharmacokinetic properties of the drug being optimized and the desired therapeutic outcomes.
Pharmacodynamic Modeling: Use mathematical models to predict drug–target interactions and optimize drug design for maximal target inhibition and minimal off-target effects.
Resistance Mechanisms
Evolutionary Dynamics Modeling: Develop mathematical models of tumor evolution to elucidate the emergence and spread of drug-resistant clones within the tumor population.
Optimal Sequencing: Determine the optimal sequence of treatments to delay or prevent the onset of resistance, considering factors such as drug mechanisms of action and resistance mechanisms.
Patient-Specific Optimization
Personalized Treatment Planning: Utilize patient-specific data, such as genetic profiles, tumor characteristics, and treatment history, to tailor treatment strategies to individual patients and optimize therapeutic outcomes.
Machine Learning Algorithms: Employ machine learning algorithms to analyze large-scale clinical data and identify predictive biomarkers of treatment response and resistance, enabling personalized treatment selection.
Here is a general mathematical formulation of the objective function:
Minimize J(θ)
where J(θ) represents the cost function or loss function, which measures the error between predicted and actual outcomes. The objective is to minimize this function to optimize the performance of the predictive model.

3.4. Feature Selection

Let X be the feature matrix representing the clinical data, where each row corresponds to a patient and each column corresponds to a feature (e.g., patient characteristics, biomarker levels, treatment history). Let y be the target variable indicating treatment response or outcome.

3.5. Algorithm Selection

Choose a suitable machine learning algorithm, such as logistic regression, decision trees, random forests, support vector machines, or deep learning models (e.g., neural networks), based on the nature of the data and the complexity of the problem.

3.6. Model Parameters

Let θ represent the parameters of the machine learning model, which are optimized during the training process to minimize the cost function J(θ).

3.7. Training

Use a training dataset to learn the parameters θ of the machine learning model. This involves optimizing the parameters to minimize the cost function using techniques such as gradient descent, stochastic gradient descent, or more advanced optimization algorithms.

3.8. Validation

After training the model, evaluate its performance on a separate validation dataset to assess its generalization ability and identify potential overfitting. This involves computing performance metrics such as accuracy, precision, recall, F1-score, and area under the receiver operating characteristic curve (AUC-ROC).

3.9. Predictive Biomarker Identification

Analyze the learned model to identify important features or biomarkers that contribute to treatment response or resistance. This can be achieved by examining the weights or coefficients of the model (e.g., for linear models) or using feature importance techniques (e.g., permutation importance, SHAP values) for tree-based models.

3.10. Personalized Treatment Selection

Once predictive biomarkers have been identified, use the trained model to predict treatment response or outcome for new patients based on their clinical data and biomarker profiles. This enables personalized treatment selection by recommending the most effective treatment strategies for individual patients [55,56].

3.11. Iterative Refinement

Iteratively refine the machine learning model by incorporating additional data, optimizing hyperparameters, or exploring different feature representations to improve predictive performance and generalization ability. By following this mathematical formulation and applying appropriate machine learning techniques, it is possible to analyze large-scale clinical data, identify predictive biomarkers of treatment response and resistance, and enable personalized treatment selection in oncology [57,58].

3.12. Resource Allocation

Use optimization techniques to evaluate the cost-effectiveness of different treatment strategies and allocate resources efficiently to maximize patient outcomes within budget constraints [57].
Healthcare Resource Optimization: Optimize the allocation of healthcare resources, such as hospital beds, medical personnel, and equipment, to ensure timely access to optimal cancer treatments for all patients. By applying mathematical optimization techniques to cancer treatments and resistance, researchers and clinicians can develop more effective and personalized therapeutic strategies to improve patient outcomes and mitigate the challenges posed by treatment resistance [58,59,60,61].

4. Application

Consider a simple mathematical model to simulate the effects of chemotherapy on tumor growth. We will use a basic exponential growth model for the tumor and incorporate a treatment effect that reduces the growth rate of tumor cells (see Figure 3).
The figure illustrates the impact of chemotherapy treatment on tumor growth over time. The blue solid line represents the exponential growth of the tumor without treatment, where tumor size increases steadily over the simulation period according to the specified growth rate. In contrast, the red dashed line represents tumor growth with chemotherapy treatment. The treatment reduces the growth rate of tumor cells, leading to a slower increase in tumor size compared to the untreated scenario. This demonstrates the efficacy of chemotherapy in inhibiting tumor progression and highlights its importance in cancer therapy. By comparing the growth curves with and without treatment, researchers can assess the effectiveness of chemotherapy in controlling tumor growth and informing treatment strategies to optimize patient outcomes.
Now, consider a more complex example involving the simulation of tumor growth and the effect of chemotherapy using a modified logistic growth model with treatment response. In this model, chemotherapy is assumed to reduce the growth rate of tumor cells over time (see Figure 4).
The figure illustrates the impact of chemotherapy treatment on tumor growth over time. The blue solid line represents the growth of the tumor without chemotherapy, following a logistic growth pattern where tumor size increases initially and then levels off due to limited resources. In contrast, the red line represents tumor growth with chemotherapy. Chemotherapy is assumed to reduce the growth rate of tumor cells, leading to slower tumor progression compared to the untreated scenario. The effect of chemotherapy becomes evident after the treatment is initiated at the specified time point, resulting in a noticeable reduction in tumor growth rate. This mathematical model provides insights into the dynamics of tumor response to chemotherapy, highlighting the potential of computational modeling to study and optimize cancer treatment strategies.
Consider a mathematical model to simulate the response of a tumor to radiation therapy. We will use a simple exponential decay model to represent the reduction in tumor size due to radiation treatment (see Figure 5).
The figure illustrates the response of a tumor to radiation therapy over a simulated time period. The blue line represents the decay of tumor size, indicating the reduction in tumor size over time due to radiation therapy. The exponential decay pattern reflects the gradual decline in tumor size as radiation treatment progresses. This simulation demonstrates the potential of mathematical modeling to study the effects of cancer treatments and optimize therapeutic strategies for improved patient outcomes.
Consider the early stages of cancer (0–20 time units). The cancer cells proliferate quickly, largely unaffected by the resistance. The rapid growth indicates an environment with plenty of resources and minimal competition or suppression (see Figure 6). As the population grows beyond initial levels, the effects of resistance become more apparent. The resistance factor (0.5) reduces the effective growth rate, indicating that biological mechanisms are impeding further growth. This stage reflects a balance between tumor cell proliferation capabilities and the body’s resistance mechanisms.
Ultimately, the growth curve reveals stabilization as the tumor approaches the carrying capacity. The model suggests that the presence of resistance contributes to a lower steady-state number of cells than would be observed in the absence of resistance, highlighting the potential impact of resistance on tumor longevity and interaction with host defenses. This example underscores the importance of understanding tumor resistance mechanisms, as they may provide critical insights for developing therapeutic strategies aimed at enhancing resistance to improve clinical outcomes in cancer treatment.
Consider a scenario where we want to optimize the parameters of a cancer treatment regime to minimize tumor growth while considering the effectiveness of a therapeutic agent in the presence of cancer resistance (see Figure 7). We want to optimize the dosage of a drug that can inhibit tumor growth while accounting for the resistance factor. The goals could include minimizing the number of cancer cells after a certain period while adhering to safety limits on drug dosage.
Here, we assume that we receive the optimal dosage of the drug required to minimize tumor proliferation while considering the effects of the resistance factor. This approach is valuable in clinical settings, as it allows for data-driven decision making regarding treatment plans. The blue curve (optimal drug dosage) represents relatively stable growth under the effective dosage, indicating that treatment manages to control or reduce tumor proliferation. The red dashed curve (no treatment) represents unchecked growth, rapidly approaching the carrying capacity, reflecting the absence of intervention. By executing the optimization procedure, we receive the optimal dosage of the drug required to minimize tumor proliferation while considering the effects of the resistance factor. This approach is valuable in clinical settings, as it allows for data-driven decision making regarding treatment plans.
This model can help in designing effective treatment regimens by balancing drug efficacy against potential resistance mechanisms present in the tumor cells. The approach can be extended to multi-drug regimens or varying resistance factors over time, incorporating patient-specific data to personalize treatment plans in oncology.
Next, we provide a compelling visualization of cancer therapy optimization. By incorporating treatment duration alongside drug dosage, we gain insights into how adjustments to therapy parameters can influence cancer cell dynamics, which is critical for developing effective therapeutic strategies (see Figure 8). Adjust the parameters further based on specific scenarios or actual experimental data for richer insights.
In the heat map representing the relationship between drug dosage (ranging from 0 to 1 in increments of 0.1), treatment duration (from 1 to 50 days in increments of 5), and the maximum cancer cell population, the intensity of color conveys critical biological insights into the effectiveness of anti-cancer treatments. For instance, at a growth rate (r) of 0.1 and a carrying capacity (K) of 1000, lighter colors at higher drug dosages (close to 1) and longer treatment durations (around 50 days) likely indicate a significantly lower maximum population of cancer cells, suggesting effective inhibition of cell growth due to the drug’s action. Conversely, darker colors may depict increased populations when either insufficient dosages or shorter durations are employed, highlighting potential resistance. This visualization allows researchers to discern optimal treatment parameters where the balance between drug efficacy and cancer cell proliferation is achieved, guiding clinical decisions for personalized cancer therapies.
In this simulation, we are examining how varying drug dosages over multiple treatment durations impact cancer cell dynamics:
  • X-axis: Represents the duration of treatment in days, highlighting different treatment intervals (e.g., 1, 6, 11, …, 51 days).
  • Y-axis: Represents varying drug dosages, from 0 to 1.
  • Color Map: The intensity of the colors illustrates the maximum cancer cell population observed. Darker colors might represent higher populations, while lighter colors indicate lower counts.
Specifically, let us consider a scenario where a drug dosage of 0.8 is applied for a duration of 40 days. If the corresponding cell on the heat map displays a light color, it would indicate that the maximum cancer cell population observed during the simulated treatment period was significantly reduced, possibly to a level below 200 cells, contingent on the color scale. In contrast, a dark cell corresponding to a low dosage of 0.2 and a short duration of 10 days could indicate that the cancer cell population peaked near the carrying capacity of 1000 cells, suggesting minimal impact of the treatment. These numerical interpretations, coupled with the visual representation, enable a nuanced understanding of the dose–response relationship and time-dependent effects of the drug on cancer cell dynamics. The results can demonstrate how combining effective drug dosages with adequate treatment duration can lead to better outcomes in controlling cancer growth.
Next, to classify the treatment response of patients, we simulated a logistic regression model that helps with understanding the relationship between patient characteristics, such as age and biomarker levels, and the likelihood of successful cancer treatment (see Figure 9). The dataset comprises two features for simplicity—age and biomarker level—represented as (X = [45, 1.5; 60, 2.1; 35, 0.8; 50, 1.2; 70, 2.5; 40, 1.0]), where each row corresponds to a patient’s age and respective biomarker level. The binary outcomes associated with these patients are captured in the vector (y = [1; 0; 1; 1; 0; 1]), indicating that the first, third, fourth, and sixth patients experienced successful treatment (success = 1), while the second and fifth patients did not (success = 0).
The decision boundary, shown as a line on the plot, represents the threshold probability of 0.5. Points on one side of the boundary are predicted to be successful treatments, while those on the opposite side are predicted to fail. The proximity of the data points to the decision boundary can also provide insights into the confidence of the model predictions, with points further from the boundary being more confidently classified. The optimized parameters (theta) obtained from the logistic regression indicate how each feature influences the treatment outcome. Optimized parameters (theta) are 8.5635, −11.8680, and −13.2920. A positive coefficient for a feature suggests that higher values of that feature increase the odds of a successful treatment response, whereas a negative coefficient suggests the opposite. In this example, the model achieved an accuracy of 100%, meaning that it correctly classified the treatment response of patients based on their features about 100% of the time. The effectiveness of the model can be improved with more comprehensive datasets, allowing it to capture more complex relationships and interactions among features.
Finally, the following example presents a logistic regression approach to assess the likelihood of patients developing resistance to antiviral treatment based on their age and viral load (see Figure 10). Logistic regression serves as a powerful tool for binary classification problems, enabling healthcare providers to predict outcomes based on patient characteristics. In this case, using patient age and viral load as predictors allows for a better understanding of how these variables contribute to the success or failure of antiviral treatment.
The model fitted using the provided data unveils patterns that suggest how increased viral loads and advanced age may correlate with resistance to treatment. By visualizing the decision boundary—where the predicted probability of a successful response is at 0.5—clinicians can identify patient profiles at higher risk for resistance. This insight can inform personalized treatment strategies, optimizing therapeutic decisions for patients facing viral infections.
The optimized parameters (theta) obtained from the logistic regression model are 8.5635, −11.8680, and −13.2920, which represent the weights assigned to the model’s features used for predicting the outcome. In this context, these parameters can be interpreted as the logarithmic odds of the predicted outcome occurring, adjusted based on the values of the corresponding features in the dataset. The first parameter is typically the intercept, while the subsequent parameters correspond to the coefficients of the respective predictors, such as patient age or viral load. With a reported model accuracy of 100%, this indicates that the model perfectly classified all instances in the training dataset with no errors, suggesting that the model may effectively distinguish between the different classes present in the data, although this could also raise concerns about overfitting, particularly if the dataset is small or lacks diversity.

5. Discussion

Mathematical modeling has become an invaluable asset in the field of oncology, providing a rigorous framework for exploring the complexities of cancer treatment, resistance mechanisms, and optimization strategies. The ability to simulate various treatment modalities allows researchers and clinicians to visualize potential outcomes under a myriad of scenarios, enhancing our understanding of how different factors interplay in the therapeutic landscape. By incorporating elements such as drug pharmacokinetics and tumor biology, models enable the prediction of treatment responses that might not be easily captured through empirical observations alone.
One of the most significant aspects addressed by mathematical modeling is the phenomenon of treatment resistance. Cancer cells often adapt to therapeutic pressures through various mechanisms, leading to treatment failure and disease progression. By utilizing models to study these resistance pathways, researchers can identify critical points for intervention, potentially guiding the development of new therapies that circumvent resistance. For instance, models can highlight the impact of genetic mutations and adaptive cellular behaviors, allowing for the design of combination therapies that work synergistically to mitigate resistance.
Moreover, the integration of optimization techniques into mathematical modeling enhances the ability to tailor treatment regimens. The personalized approach to cancer therapy is a cornerstone of modern oncology, aiming to maximize effectiveness while minimizing toxicity. Through optimization models, clinicians can develop individualized treatment plans based on patient-specific data, such as tumor characteristics and treatment history. This personalized approach not only has the potential to improve patient outcomes but also to reduce healthcare costs by avoiding ineffective treatment strategies.
While the potential of mathematical modeling in cancer therapy is immense, it is crucial to acknowledge its limitations. Models depend heavily on the accuracy and completeness of input data, which can sometimes be lacking or variable across different patient populations. Additionally, the complexity of biological systems means that no model can capture every nuance of tumor behavior or patient response. Therefore, continuous validation against clinical data and iterative refinement of models will be essential for maintaining their relevance and applicability in clinical practice.
Future work will focus on validating the model’s predictions against clinical datasets and experimental results to refine its accuracy and applicability to real-world cancer treatment scenarios. This will involve estimating key model parameters from patient data and comparing predicted tumor growth dynamics with observed clinical outcomes. Furthermore, while our model provides a valuable framework for understanding tumor dynamics, it is crucial to acknowledge that its predictive power is inherently limited by the quality and potential biases present in the input data, as well as the exclusion of complex biological factors such as microenvironmental influences, immune responses, and intratumoral heterogeneity. Future iterations of the model will aim to incorporate these complexities and address data bias to improve its predictive capabilities and clinical relevance.

6. Conclusions

In conclusion, mathematical modeling serves as a powerful tool in understanding and optimizing cancer treatments. By simulating the dynamics of treatment responses, elucidating resistance mechanisms, and applying optimization techniques for personalized therapy, mathematical models bring a systematic approach to decision making in oncology. As research accelerates and computational capabilities expand, the integration of mathematical modeling with experimental and clinical data will further enhance our ability to develop effective and personalized cancer treatments. Moving forward, ongoing collaboration between mathematicians, biologists, and clinicians will be key to overcoming the challenges inherent in cancer therapies and ultimately improving patient prognoses. Embracing the insights provided by these models can significantly contribute to the evolution of cancer treatment paradigms, paving the way for innovative and effective therapeutic strategies in the future.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Sun, X.; Hu, B. Mathematical modeling and computational prediction of cancer drug resistance. Brief. Bioinform. 2018, 19, 1382–1399. [Google Scholar] [CrossRef] [PubMed]
  2. Swierniak, A.; Kimmel, M.; Smieja, J. Mathematical modeling as a tool for planning anticancer therapy. Eur. J. Pharmacol. 2009, 625, 108–121. [Google Scholar] [CrossRef] [PubMed]
  3. Schättler, H.; Ledzewicz, U. Optimal control for mathematical models of cancer therapies. In An Application of Geometric Methods; Springer: Cham, Switzerland, 2015. [Google Scholar]
  4. Li, L.; Zhao, T.; Hu, Y.; Ren, S.; Tian, T. Mathematical modelling and bioinformatics analyses of drug resistance for cancer treatment. Curr. Bioinform. 2024, 19, 211–221. [Google Scholar]
  5. Azizi, T. Mathematical Modeling with Applications in Biological Systems, Physiology, and Neuroscience; Kansas State University: Manhattan, KS, USA, 2021. [Google Scholar]
  6. Azizi, T.; Alali, B.; Kerr, G. Mathematical Modeling: With Applications in Physics, Biology, Chemistry, and Engineering, 2nd ed.; BP International: London, UK, 2021. [Google Scholar]
  7. Azizi, T. Mathematical Modeling of Cancer Progression. AppliedMath 2024, 4, 1065–1079. [Google Scholar] [CrossRef]
  8. Lytle, N.K.; Barber, A.G.; Reya, T. Stem cell fate in cancer growth, progression and therapy resistance. Nat. Rev. Cancer 2018, 18, 669–680. [Google Scholar] [CrossRef]
  9. Wang, X.; Zhang, H.; Chen, X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019, 2, 141. [Google Scholar] [CrossRef]
  10. Rebucci, M.; Michiels, C. Molecular aspects of cancer cell resistance to chemotherapy. Biochem. Pharmacol. 2013, 85, 1219–1226. [Google Scholar] [CrossRef]
  11. Baquero, F.; Martinez, J.L.; FLanza, V.; Rodríguez-Beltrán, J.; Galán, J.C.; San Millán, A.; Cantón, R.; Coque, T.M. Evolutionary pathways and trajectories in antibiotic resistance. Clin. Microbiol. Rev. 2021, 34, e00050-19. [Google Scholar] [CrossRef]
  12. Azizi, T. Intersections of Neurobiology and Oncology: Foundations and Frontiers in Cancer Neuroscience; BP International: London, UK, 2024. [Google Scholar]
  13. Craig, M.; Jenner, A.L.; Namgung, B.; Lee, L.P.; Goldman, A. Engineering in medicine to address the challenge of cancer drug resistance: From micro-and nanotechnologies to computational and mathematical modeling. Chem. Rev. 2020, 121, 3352–3389. [Google Scholar] [CrossRef]
  14. Kulesza, A.; Couty, C.; Lemarre, P.; Thalhauser, C.J.; Cao, Y. Advancing cancer drug development with mechanistic mathematical modeling: Bridging the gap between theory and practice. J. Pharmacokinet. Pharmacodyn. 2024, 51, 581–604. [Google Scholar] [CrossRef]
  15. Beerenwinkel, N.; Schwarz, R.F.; Gerstung, M.; Markowetz, F. Cancer evolution: Mathematical models and computational inference. Syst. Biol. 2015, 64, e1–e25. [Google Scholar] [CrossRef] [PubMed]
  16. Barbolosi, D.; Ciccolini, J.; Lacarelle, B.; Barlési, F.; André, N. Computational oncology—Mathematical modelling of drug regimens for precision medicine. Nat. Rev. Clin. Oncol. 2016, 13, 242–254. [Google Scholar] [CrossRef] [PubMed]
  17. Ferrer, F.; Fanciullino, R.; Milano, G.; Ciccolini, J. Towards rational cancer therapeutics: Optimizing dosing, delivery, scheduling, and combinations. Clin. Pharmacol. Ther. 2020, 108, 458–470. [Google Scholar] [CrossRef] [PubMed]
  18. McDonald, T.O.; Cheng, Y.C.; Graser, C.; Nicol, P.B.; Temko, D.; Michor, F. Computational approaches to modelling and optimizing cancer treatment. Nat. Rev. Bioeng. 2023, 1, 695–711. [Google Scholar] [CrossRef]
  19. DeRidder, L.; Rubinson, D.A.; Langer, R.; Traverso, G. The past, present, and future of chemotherapy with a focus on individualization of drug dosing. J. Control. Release 2022, 352, 840–860. [Google Scholar] [CrossRef]
  20. Malinzi, J.; Basita, K.B.; Padidar, S.; Adeola, H.A. Prospect for application of mathematical models in combination cancer treatments. Inform. Med. Unlocked 2021, 23, 100534. [Google Scholar] [CrossRef]
  21. Yin, A.; Moes, D.J.A.; van Hasselt, J.G.; Swen, J.J.; Guchelaar, H.J. A review of mathematical models for tumor dynamics and treatment resistance evolution of solid tumors. CPT Pharmacomet. Syst. Pharmacol. 2019, 8, 720–737. [Google Scholar] [CrossRef]
  22. Sun, X.; Bao, J.; Shao, Y. Mathematical modeling of therapy-induced cancer drug resistance: Connecting cancer mechanisms to population survival rates. Sci. Rep. 2016, 6, 22498. [Google Scholar] [CrossRef]
  23. Olushola, A.; Alao, V. Biomathematics in Cancer Research: Looking into How Mathematical Models Are Used to Understand Tumor Growth and the Effectiveness of Different Treatment Strategies. 2024; preprint. [Google Scholar]
  24. Padmanabhan, R.; Meskin, N.; Al Moustafa, A.E. Mathematical Models of Cancer and Different Therapies; Springer: Singapore, 2021; pp. 135–156. [Google Scholar]
  25. Altrock, P.M.; Liu, L.L.; Michor, F. The mathematics of cancer: Integrating quantitative models. Nat. Rev. Cancer 2015, 15, 730–745. [Google Scholar] [CrossRef]
  26. Dogra, P.; Ramirez, J.R.; Peláez, M.J.; Wang, Z.; Cristini, V.; Parasher, G.; Rawat, M. Mathematical modeling to address challenges in pancreatic cancer. Curr. Top. Med. Chem. 2020, 20, 367–376. [Google Scholar] [CrossRef]
  27. Benzekry, S.; Lamont, C.; Beheshti, A.; Tracz, A.; Ebos, J.M.; Hlatky, L.; Hahnfeldt, P. Classical mathematical models for description and prediction of experimental tumor growth. PLoS Comput. Biol. 2014, 10, e1003800. [Google Scholar]
  28. Sheergojri, A.R.; Iqbal, P.; Agarwal, P.; Özdemir, N. Uncertainty-based Gompertz growth model for tumor population and its numerical analysis. Int. J. Optim. Control-Theor. Appl. 2022, 12, 137–150. [Google Scholar]
  29. Azizi, T.; Robert, M. Global sensitivity analysis in physiological systems. Appl. Math. 2020, 11, 119–136. [Google Scholar]
  30. Abdulrashid, I.; Delen, D.; Usman, B.; Uzochukwu, M.I.; Ahmed, I. A multi-objective optimization framework for determining optimal chemotherapy dosing and treatment duration. Healthc. Anal. 2024, 5, 100335. [Google Scholar] [CrossRef]
  31. Jarrett, A.M.; Faghihi, D.; Hormuth, D.A.; Lima, E.A.; Virostko, J.; Biros, G.; Patt, D.; Yankeelov, T.E. Optimal control theory for personalized therapeutic regimens in oncology: Background, history, challenges, and opportunities. J. Clin. Med. 2020, 9, 1314. [Google Scholar] [CrossRef]
  32. Hassan, M.; Awan, F.M.; Naz, A.; deAndrés-Galiana, E.J.; Alvarez, O.; Cernea, A.; Fernández-Brillet, L.; Fernández-Martínez, J.L.; Kloczkowski, A. Innovations in genomics and big data analytics for personalized medicine and health care: A review. Int. J. Mol. Sci. 2022, 23, 4645. [Google Scholar] [CrossRef]
  33. Ho, D.; Quake, S.R.; McCabe, E.R.; Chng, W.J.; Chow, E.K.; Ding, X.; Gelb, B.D.; Ginsburg, G.S.; Hassenstab, J.; Ho, C.M.; et al. Enabling technologies for personalized and precision medicine. Trends Biotechnol. 2020, 38, 497–518. [Google Scholar]
  34. Krzyszczyk, P.; Acevedo, A.; Davidoff, E.J.; Timmins, L.M.; Marrero-Berrios, I.; Patel, M.; White, C.; Lowe, C.; Sherba, J.J.; Hartmanshenn, C.; et al. The growing role of precision and personalized medicine for cancer treatment. Technology 2018, 6, 79–100. [Google Scholar]
  35. Gatenby, R.A.; Brown, J.S. The evolution and ecology of resistance in cancer therapy. Cold Spring Harb. Perspect. Med. 2020, 10, a040972. [Google Scholar] [CrossRef]
  36. Butner, J.D.; Dogra, P.; Chung, C.; Pasqualini, R.; Arap, W.; Lowengrub, J.; Cristini, V.; Wang, Z. Mathematical modeling of cancer immunotherapy for personalized clinical translation. Nat. Comput. Sci. 2022, 2, 785–796. [Google Scholar]
  37. Schlicke, P.; Kuttler, C.; Schumann, C. How mathematical modeling could contribute to the quantification of metastatic tumor burden under therapy: Insights in immunotherapeutic treatment of non-small cell lung cancer. Theor. Biol. Med. Model. 2021, 18, 11. [Google Scholar]
  38. Gatenby, R.A.; Brown, J.S. Integrating evolutionary dynamics into cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 675–686. [Google Scholar] [PubMed]
  39. Moharamipour, S.; Aminifar, M.; Foroughi-Gilvaee, M.R.; Faranoush, P.; Mahdavi, R.; Abadijoo, H.; Parniani, M.; Abbasvandi, F.; Mansouri, S.; Abdolahad, M. Hydroelectric actuator for 3-dimensional analysis of electrophoretic and dielectrophoretic behavior of cancer cells; suitable in diagnosis and invasion studies. Biomater. Adv. 2023, 151, 213476. [Google Scholar]
  40. Zahir, N.; Sun, R.; Gallahan, D.; Gatenby, R.A.; Curtis, C. Characterizing the ecological and evolutionary dynamics of cancer. Nat. Genet. 2020, 52, 759–767. [Google Scholar]
  41. Kartal-Yandim, M.; Adan-Gokbulut, A.; Baran, Y. Molecular mechanisms of drug resistance and its reversal in cancer. Crit. Rev. Biotechnol. 2016, 36, 716–726. [Google Scholar]
  42. Casadei, B.; Giacosa, M.; Maula, A.; Plos, S.; Zappulla, L.; Viotto, C.; Deriu, M.A.; Tuszynski, J.A. Complexities of drug resistance in cancer: An overview of strategies and mathematical models. In Cancer, Complexity, Computation; Springer: Cham, Switzerland, 2022; pp. 309–332. [Google Scholar]
  43. Xie, H.; Jiao, Y.; Fan, Q.; Hai, M.; Yang, J.; Hu, Z.; Yang, Y.; Shuai, J.; Chen, G.; Liu, R.; et al. Modeling three-dimensional invasive solid tumor growth in heterogeneous microenvironment under chemotherapy. PLoS ONE 2018, 13, e0206292. [Google Scholar]
  44. Norton, K.A.; Gong, C.; Jamalian, S.; Popel, A.S. Multiscale agent-based and hybrid modeling of the tumor immune microenvironment. Processes 2019, 7, 37. [Google Scholar] [CrossRef]
  45. Rockne, R.C.; Hawkins-Daarud, A.; Swanson, K.R.; Sluka, J.P.; Glazier, J.A.; Macklin, P.; Hormuth, D.A.; Jarrett, A.M.; Lima, E.A.; Oden, J.T.; et al. The 2019 mathematical oncology roadmap. Phys. Biol. 2019, 16, 041005. [Google Scholar]
  46. Hoffmann, K.; Cazemier, K.; Baldow, C.; Schuster, S.; Kheifetz, Y.; Schirm, S.; Horn, M.; Ernst, T.; Volgmann, C.; Thiede, C.; et al. Integration of mathematical model predictions into routine workflows to support clinical decision making in haematology. BMC Med. Inform. Decis. Mak. 2020, 20, 1–12. [Google Scholar]
  47. Msaouel, P.; Lee, J.; Thall, P.F. Making patient-specific treatment decisions using prognostic variables and utilities of clinical outcomes. Cancers 2021, 13, 2741. [Google Scholar] [CrossRef]
  48. Grassberger, C.; McClatchy, D., III; Geng, C.; Kamran, S.C.; Fintelmann, F.; Maruvka, Y.E.; Piotrowska, Z.; Willers, H.; Sequist, L.V.; Hata, A.N.; et al. Patient-specific tumor growth trajectories determine persistent and resistant cancer cell populations during treatment with targeted therapies. Cancer Res. 2019, 79, 3776–3788. [Google Scholar] [PubMed]
  49. Johnson, K.E.; Howard, G.R.; Morgan, D.; Brenner, E.A.; Gardner, A.L.; Durrett, R.E.; Mo, W.; Al’Khafaji, A.; Sontag, E.D.; Jarrett, A.M.; et al. Integrating transcriptomics and bulk time course data into a mathematical framework to describe and predict therapeutic resistance in cancer. Phys. Biol. 2020, 18, 016001. [Google Scholar]
  50. Cunningham, J.J.; Brown, J.S.; Gatenby, R.A.; Staňková, K. Optimal control to develop therapeutic strategies for metastatic castrate resistant prostate cancer. J. Theor. Biol. 2018, 459, 67–78. [Google Scholar] [PubMed]
  51. Bräutigam, K. Optimization of chemotherapy regimens using mathematical programming. Comput. Ind. Eng. 2024, 191, 110078. [Google Scholar]
  52. Ribba, B.; Boetsch, C.; Nayak, T.; Grimm, H.P.; Charo, J.; Evers, S.; Klein, C.; Tessier, J.; Charoin, J.E.; Phipps, A.; et al. Prediction of the optimal dosing regimen using a mathematical model of tumor uptake for immunocytokine-based cancer immunotherapy. Clin. Cancer Res. 2018, 24, 3325–3333. [Google Scholar]
  53. Wen, H.; Jung, H.; Li, X. Drug delivery approaches in addressing clinical pharmacology-related issues: Opportunities and challenges. AAPS J. 2015, 17, 1327–1340. [Google Scholar]
  54. Setia, A.; Sahu, R.K.; Ray, S.; Widyowati, R.; Ekasari, W.; Saraf, S. Advances in hybrid vesicular-based drug delivery systems: Improved biocompatibility, targeting, therapeutic efficacy and pharmacokinetics of anticancer drugs. Curr. Drug Metab. 2022, 23, 757–780. [Google Scholar]
  55. Trebeschi, S.; Drago, S.G.; Birkbak, N.J.; Kurilova, I.; Cǎlin, A.M.; Pizzi, A.D.; Lalezari, F.; Lambregts, D.M.J.; Rohaan, M.W.; Parmar, C.; et al. Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann. Oncol. 2019, 30, 998–1004. [Google Scholar]
  56. Adam, G.; Rampášek, L.; Safikhani, Z.; Smirnov, P.; Haibe-Kains, B.; Goldenberg, A. Machine learning approaches to drug response prediction: Challenges and recent progress. NPJ Precis. Oncol. 2020, 4, 19. [Google Scholar]
  57. Ippolito, P.P. Hyperparameter Tuning: The Art of Fine-Tuning Machine and Deep Learning Models to Improve Metric Results. In Applied Data Science in Tourism: Interdisciplinary Approaches, Methodologies, and Applications; Springer International Publishing: Cham, Switzerland, 2022; pp. 231–251. [Google Scholar]
  58. Waring, J.; Lindvall, C.; Umeton, R. Automated machine learning: Review of the state-of-the-art and opportunities for healthcare. Artif. Intell. Med. 2020, 104, 101822. [Google Scholar]
  59. Crown, W.; Buyukkaramikli, N.; Thokala, P.; Morton, A.; Sir, M.Y.; Marshall, D.A.; Tosh, J.; Padula, W.V.; Ijzerman, M.J.; Wong, P.K.; et al. Constrained optimization methods in health services research—An introduction: Report 1 of the ISPOR optimization methods emerging good practices task force. Value Health 2017, 20, 310–319. [Google Scholar] [PubMed]
  60. Mansour, R.; Abdel-Razeq, H.; Al-Hussaini, M.; Shamieh, O.; Al-Ibraheem, A.; Al-Omari, A.; Mansour, A.H. Systemic Barriers to Optimal Cancer Care in Resource-Limited Countries: Jordanian Healthcare as an Example. Cancers 2024, 16, 1117. [Google Scholar] [CrossRef] [PubMed]
  61. Saxena, A.K.; Dixit, R.R.; Aman-Ullah, A. An LSTM Neural Network Approach to Resource Allocation in Hospital Management Systems. Int. J. Appl. Health Care Anal. 2022, 7, 1–12. [Google Scholar]
Figure 1. Schematic diagram of different types of cancer treatment (created in https://BioRender.com).
Figure 1. Schematic diagram of different types of cancer treatment (created in https://BioRender.com).
Appliedmath 05 00040 g001
Figure 2. Workflow of the study on mathematical modeling of cancer treatment.
Figure 2. Workflow of the study on mathematical modeling of cancer treatment.
Appliedmath 05 00040 g002
Figure 3. Simulation of tumor growth with and without chemotherapy treatment using a mathematical model. The blue solid line represents tumor growth without treatment, while the red dashed line indicates tumor growth with chemotherapy. Treatment reduces the growth rate of tumor cells, resulting in slower tumor progression.
Figure 3. Simulation of tumor growth with and without chemotherapy treatment using a mathematical model. The blue solid line represents tumor growth without treatment, while the red dashed line indicates tumor growth with chemotherapy. Treatment reduces the growth rate of tumor cells, resulting in slower tumor progression.
Appliedmath 05 00040 g003
Figure 4. Simulation of tumor growth with and without chemotherapy treatment using a modified logistic growth model. The blue solid line represents tumor growth without chemotherapy, while the red line indicates tumor growth with chemotherapy. Chemotherapy reduces the growth rate of tumor cells, resulting in slower tumor progression after treatment initiation.
Figure 4. Simulation of tumor growth with and without chemotherapy treatment using a modified logistic growth model. The blue solid line represents tumor growth without chemotherapy, while the red line indicates tumor growth with chemotherapy. Chemotherapy reduces the growth rate of tumor cells, resulting in slower tumor progression after treatment initiation.
Appliedmath 05 00040 g004
Figure 5. Simulation of tumor response to radiation therapy using a mathematical model. The red line represents the decay of tumor size over time due to radiation therapy, the blue line represents the exponential growth of tumor size without radiation therapy.
Figure 5. Simulation of tumor response to radiation therapy using a mathematical model. The red line represents the decay of tumor size over time due to radiation therapy, the blue line represents the exponential growth of tumor size without radiation therapy.
Appliedmath 05 00040 g005
Figure 6. Simulation of cancer cell growth dynamics under the influence of a resistance factor. The graph depicts the number of cancer cells over time, starting with an initial population of 10 cells. The intrinsic growth rate is set at 0.1, and the carrying capacity of the environment is 1000 cells. The resistance factor valued at 0.5 indicates a moderate level of cancer resistance, which effectively reduces the growth rate of the tumor. As time progresses, the growth curve demonstrates a logistic pattern influenced by the resistance, suggesting that resistance mechanisms can significantly impact cancer proliferation, potentially leading to prolonged survivability and stability of the tumor population.
Figure 6. Simulation of cancer cell growth dynamics under the influence of a resistance factor. The graph depicts the number of cancer cells over time, starting with an initial population of 10 cells. The intrinsic growth rate is set at 0.1, and the carrying capacity of the environment is 1000 cells. The resistance factor valued at 0.5 indicates a moderate level of cancer resistance, which effectively reduces the growth rate of the tumor. As time progresses, the growth curve demonstrates a logistic pattern influenced by the resistance, suggesting that resistance mechanisms can significantly impact cancer proliferation, potentially leading to prolonged survivability and stability of the tumor population.
Appliedmath 05 00040 g006
Figure 7. Effects of optimized drug dosage on cancer cell proliferation. This plot illustrates the evolution of the cancer cell population over time under two scenarios: (1) treatment with the optimized drug dosage (blue solid line) and (2) the absence of treatment (red dashed line). The optimized dosage effectively reduces cancer cell growth compared to the untreated scenario, where the population rapidly approaches its carrying capacity. These results highlight the potential of tailored therapeutic interventions in managing tumor growth while accounting for resistance mechanisms.
Figure 7. Effects of optimized drug dosage on cancer cell proliferation. This plot illustrates the evolution of the cancer cell population over time under two scenarios: (1) treatment with the optimized drug dosage (blue solid line) and (2) the absence of treatment (red dashed line). The optimized dosage effectively reduces cancer cell growth compared to the untreated scenario, where the population rapidly approaches its carrying capacity. These results highlight the potential of tailored therapeutic interventions in managing tumor growth while accounting for resistance mechanisms.
Appliedmath 05 00040 g007
Figure 8. Heat map of maximum cancer cell population in response to drug dosage and treatment duration. This heat map illustrates the interaction between varying drug dosages and treatment durations on the maximum population of cancer cells over a treatment period of up to 100 days. The x-axis shows the duration of therapy, while the y-axis displays the range of drug dosages administered. The color gradient indicates the maximum observed cancer cell population during the corresponding treatment conditions, with darker colors reflecting higher populations. This visualization highlights the significance of optimizing both drug dosage and treatment duration in achieving effective cancer control, suggesting that the right combination can significantly reduce tumor growth.
Figure 8. Heat map of maximum cancer cell population in response to drug dosage and treatment duration. This heat map illustrates the interaction between varying drug dosages and treatment durations on the maximum population of cancer cells over a treatment period of up to 100 days. The x-axis shows the duration of therapy, while the y-axis displays the range of drug dosages administered. The color gradient indicates the maximum observed cancer cell population during the corresponding treatment conditions, with darker colors reflecting higher populations. This visualization highlights the significance of optimizing both drug dosage and treatment duration in achieving effective cancer control, suggesting that the right combination can significantly reduce tumor growth.
Appliedmath 05 00040 g008
Figure 9. Visualization of logistic regression model for predicting cancer treatment response. The scatter plot illustrates the normalized values of age and biomarker levels for patients, differentiated by treatment outcomes: success (1) indicated by red circles and failure (0) marked by blue crosses. The black contour line represents the decision boundary where the predicted probability of treatment success is 0.5, effectively separating the two classes based on the logistic regression model. This visualization aids in understanding how the model classifies patients and the features that contribute to the likelihood of treatment success.
Figure 9. Visualization of logistic regression model for predicting cancer treatment response. The scatter plot illustrates the normalized values of age and biomarker levels for patients, differentiated by treatment outcomes: success (1) indicated by red circles and failure (0) marked by blue crosses. The black contour line represents the decision boundary where the predicted probability of treatment success is 0.5, effectively separating the two classes based on the logistic regression model. This visualization aids in understanding how the model classifies patients and the features that contribute to the likelihood of treatment success.
Appliedmath 05 00040 g009
Figure 10. Visualization of logistic regression model for predicting chemotherapy resistance in cancer patients. This scatter plot illustrates the relationship between patient age and tumor size, categorizing treatment outcomes as successful responses (red circles) or resistance (blue crosses). The dashed black line indicates the decision boundary established by the logistic regression model, separating patients likely to respond favorably to chemotherapy from those predicted to have resistance.
Figure 10. Visualization of logistic regression model for predicting chemotherapy resistance in cancer patients. This scatter plot illustrates the relationship between patient age and tumor size, categorizing treatment outcomes as successful responses (red circles) or resistance (blue crosses). The dashed black line indicates the decision boundary established by the logistic regression model, separating patients likely to respond favorably to chemotherapy from those predicted to have resistance.
Appliedmath 05 00040 g010
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Azizi, T. Mathematical Modelling of Cancer Treatments, Resistance, Optimization. AppliedMath 2025, 5, 40. https://doi.org/10.3390/appliedmath5020040

AMA Style

Azizi T. Mathematical Modelling of Cancer Treatments, Resistance, Optimization. AppliedMath. 2025; 5(2):40. https://doi.org/10.3390/appliedmath5020040

Chicago/Turabian Style

Azizi, Tahmineh. 2025. "Mathematical Modelling of Cancer Treatments, Resistance, Optimization" AppliedMath 5, no. 2: 40. https://doi.org/10.3390/appliedmath5020040

APA Style

Azizi, T. (2025). Mathematical Modelling of Cancer Treatments, Resistance, Optimization. AppliedMath, 5(2), 40. https://doi.org/10.3390/appliedmath5020040

Article Metrics

Back to TopTop