1. Introduction
The versatility of mathematical and computational models has made them an increasingly crucial tool in biomedical research. Models create abstract and simplified representations of real-world phenomena, allowing researchers to gain deeper insights into inherently complex biological processes. These biologically driven and carefully calibrated models extend beyond purely theoretical pursuits. They can shed light on important underlying mechanisms, predict emergent patterns [
1], test therapeutic strategies [
2], and even inform the design of clinical trials [
3,
4]. Over the past decade, immunotherapy has established a new paradigm for the treatment of cancer [
5,
6]. Immunotherapy is fundamentally different from traditional first-line therapies, such as radiation and chemotherapy [
7]. By harnessing the immune system’s power, immunotherapy overcomes immunosuppression induced by a tumor and its microenvironment, allowing the immune system to target and kill cancer cells [
5,
6]. Among the various immunotherapy methods such as direct immune modulators, monoclonal antibodies, oncolytic viruses, adoptive cell therapy and vaccines [
6,
8,
9], immune checkpoint inhibitors (ICIs) have garnered significant attention. ICIs are a class of immunotherapeutics that reinvigorate the killing activities of immune cells by blocking the activation of inhibitory immunoreceptors [
7,
10]. They have shown remarkable results for many patients. However, ICI monotherapy’s low overall response rates and difficulty enhancing patients’ responses with combination therapy in many cancers present an ongoing challenge to clinicians [
8,
11,
12].
ICIs aim to revitalize cytotoxic T lymphocytes (CTLs), which are a key component of the adaptive immune system and major killers of pathogens and neoplastic cells [
13,
14,
15,
16]. Adding further complexity to the varied antitumor immune responses is the fact that cytotoxic T lymphocytes (CTLs) execute their cell-killing function via at least two distinct mechanisms [
17,
18]. The first process is mediated by perforin and granzymes. Perforin facilitates the formation of pores in the target cell membrane, which allows granzymes to access the target cell cytoplasm to induce apoptosis [
17,
19,
20]. The second process is through the Fas pathway. FasL, a type II transmembrane protein upregulated on CTLs, can engage Fas on the target cell to trigger apoptosis of the target cell [
17,
21]. Evidence showed that the perforin/granzyme-mediated process happens faster than the FasL-mediated process [
17]. In an in vitro study, perforin-mediated killing was completed within thirty minutes, whereas FasL-based killing was detected no sooner than two hours after the tumor cell was conjugated with CTL [
21]. Evidence also showed that the switch from fast to slow killing is related to the decreasing presence of antigens [
22]. Although the connections between distinct CTL killing mechanisms are not fully understood, we find it important to consider the immune system’s varied responses towards tumor cells with different antigenicity and to integrate them into our computational models.
With the increasing amount of high-throughput data to analyze and the plethora of treatment strategies to test, reliable and cost-efficient computational modeling becomes an indispensable tool in studies of cancer immunotherapy [
23,
24]. To explain the wide variations of patient responses, quantify the influence of spatial complexity in the tumor microenvironment (TME), and predict which patients are most likely to respond well to ICIs, we build mathematical and computational models for the ICIs targeting the PD-1/PD-L1 immune checkpoint. Differential equation-based models and agent-based models (ABMs) are popular modeling approaches for cancer treatments. Ordinary differential equation (ODE) models describe the temporal evolution of populations of cells or molecules through a set of coupled mathematical equations. In contrast, partial differential equation (PDE) models describe spatial-temporal dynamics using densities of cells or concentration gradients. ODE-based models in immuno-oncology include those that represent general tumor-immune dynamics [
25], oncolytic virus therapy [
26], anti-PD-1 immune checkpoint inhibitors [
27], resistance to dendritic-cell vaccines [
28] and so on. PDE-based models include but are not limited to [
29,
30], which focus on therapies involving anti-PD1 ICIs. On the other hand, an ABM simulates how individual entities, such as cells and molecules, move and interact with each other and with the environment. Many ABMs have been developed to model the TME and cancer immune response [
31]. We previously developed the first ODE model building on the works of [
25,
27], and subsequently the first ABM for anti-PD-1 immune checkpoint blockade therapy with consideration of tumor cells of different antigenicity and the two aforementioned CTL killing mechanisms [
32,
33]. However, the ABM in [
33] also includes the anti-FGFR3 small molecule inhibitors. The ABM in this paper is adapted from [
33] to focus on the activity of the PD-1/PD-L1 immune checkpoint and the two CTL killing mechanisms like in the ODE model.
By comparing fundamentally different modeling approaches, an ODE model, and an ABM of the same biological process, we make use of their strengths and also explore their limitations. ODEs allow rapid simulations and thorough diagnostics. On the other hand, ABMs reflect the discrete nature of biology better [
24] and can reveal emergent behaviors that would be missed in a purely equation-based approach. While our previous work analyzed the ODE model in detail to identify important characteristics of the tumor-immune landscape that have the largest impact on the outcomes of immune checkpoint blockade, this paper centers on examining what aspects of the tumor-immune dynamics both the ODEs and ABM can describe and what unique insights the ABM can offer due to the integration of the spatial elements. By comparing and contrasting the ABM and the ODE model and using the immune checkpoint inhibitors as an example, we will also discuss the balance between model tractability, model complexity, and computational efficiency when building models for cancer immunotherapy. The paper is structured as follows:
Section 2 explains our experimental and modeling methods;
Section 3 provides a detailed description of the simulation results, which serve as the basis for the discussions in
Section 4 on the preclinical and clinical implications of our models and a comparison of the two modeling strategies.
Section 5 concludes the paper.
2. Materials and Methods
Key steps of our experimental, modeling, and analytical pipeline are outlined in
Figure 1. We use in vivo data to build a biologically informed ODE model and ABM to simulate virtual tumors in a virtual cohort with diverse tumor-immune characteristics and predict individual responses to immune checkpoint blockade therapy. Further details about the experiments, model formulation, and model calibration are provided in this section.
2.1. Computational Models
We compare two mathematical models to describe the tumor-immune dynamics with an active or blocked PD1/PD-L1 immune checkpoint. The first formulation is an ODE model that tracks the temporal changes in the number of tumor cells, CTLs, and concentration of PD-1 and PD-L1. The details of this ODE model are previously published in [
32]. The second formulation is a three-dimensional, on-lattice ABM in which tumor cells and immune cells are modeled as autonomous agents interacting with each other and the TME. Like in the ODE model, the ABM has three types of cells: high-antigen (HA) tumor cells, low-antigen (LA) tumor cells, and CTLs. Cells in the ABM occupy lattice sites. Tumor cells are immobile, while CTLs are mobile. At each time step, tumor cells can proliferate or undergo apoptosis. The proliferation of tumor cells slows down due to contact inhibition [
34] because tumor cells are immobile in this ABM. Here, we only simulate the virtual tumor until it escapes or metastasizes into nearby blood vessels. Hence, simulations stop when the tumor cells exceed the maximum number allowed or too many tumor cells have reached the boundaries of the TME lattice. The model employs an immune stimulatory factor (ISF), a construct representing the combined effect of factors that each tumor cell secretes into the local neighborhood of the tumor microenvironment. The level of ISF expression depends on the cell’s antigenicity. LA tumor cells secrete a fraction of ISF compared to HA tumor cells.
In the ABM, CTLs are recruited from the lattice boundaries at a constant rate, independent of tumor size. At each time step, a CTL can execute one of the following actions: proliferation, apoptosis or exhaustion, movement, or conjugation. The proliferation rate of CTLs depends on both a base rate and the concentration of ISFs in the surrounding environment and is also affected by contact inhibition. CTL exhaustion occurs as a result of extended antigen exposure [
35,
36,
37]. CTL apoptosis also arises naturally [
37]. Since both dead and exhausted CTLs lose effector functions, the apoptosis and exhaustion of CTLs are combined into a single event in the ABM. The direction of CTL movement is influenced by the concentration gradient of ISF in the TME, i.e., CTLs are more likely to move in the direction of higher ISF. Once CTLs conjugate with a tumor cell, they attempt to destroy it via fast or slow killing. In our previous ABM [
33], HA tumor cells are only killed via the fast mechanism, and the LA tumor cells are only killed via the slow mechanism. We relax this restriction, adding the probability of fast killing for both HA and LA tumor cells. This allows maximum modeling flexibility and allows us to assess the importance of considering the two killing mechanisms in tumor-immune dynamics. The assumption in the baseline parameter set is that CTLs kill HA tumor cells preferentially via the fast mechanism and kill LA tumor cells preferentially via the slow mechanism. In both the ABM and the ODE model, an active PD-1/PD-L1 immune checkpoint inhibits the recruitment and antigen-mediated proliferation rates of CTLs in the TME. In both models, we categorize therapeutic outcomes into “elimination”, “dormancy”, and “escape”, which correspond to the three phases of immunoediting: elimination, equilibrium, and escape [
38,
39].
2.2. Description of Experiments
For mouse experiments, 6–8 week old female RAG1 KO and C57BL/6J mice were obtained from The Jackson Laboratory. Mice were housed in a specific pathogen-free animal facility at the University of Chicago and used in accordance with the animal experimental guidelines set by the University of Chicago Animal Care and Use Committee (IACUC). All experimental animal procedures were approved by the IACUC.
The MB49 cell line is a chemical carcinogen-induced urothelial carcinoma cell line derived from a male C57BL/lcrf-a’ mouse. Cells were maintained at 37 °C with 5% CO2 in DMEM supplemented with 10% heat-inactivated FCS, penicillin, and streptomycin. MB49 tumor cells were subcutaneously injected into the flank of RAG1 KO (n = 27) or C57Bl/6J (n = 24). Four types of MB49 cells with different expression levels of the model antigen SIY (SIYRYYGL) were used: Zs green (no SIY), L14 (low SIY), H1 (high SIY), and a mix of L14 and H1 cells with 1:1 ratio. Each type of MB49 cell were injected into five to seven mice of each strain. Mice that died or had tumors with more than 50% ulceration were excluded from the data used for model calibration. On Days 7, 10, 12, 14, 17, and 19, tumors were measured three-dimensionally using a digital caliper. Tumor volume was calculated using L × W × H. All mice were sacrificed on Day 20 in accordance with IACUC guidelines for humane endpoints. Tumors were harvested and digested in 10% FBS/RPMI. Single cell suspensions were filtered through a 100 M cell strainer and stained with antibodies to PD-1, CD69, CD3, CD19, LAG3, Ki67, CD4, CD44, CD45, CD8a, SIY, CD62L, Foxp3, and Live/Dead Viability Dye Zombie NIR. CTLs were analyzed using flow cytometry. The number of CD8 cells is directly measured and the CTL density within the tumor, i.e., the number of CD8 cells per of tumor is calculated.
2.3. Estimation of Model Parameters and Construction of Virtual Tumors
To convert tumor volume to number of tumor cells, we assume that 1
of tumor is equivalent to
tumor cells [
40]. Given this conversion rate and the initial conditions of the experiments and the ABM simulations, each ABM tumor cell represents 50,000 actual tumor cells. The proliferation rate (
) and the carrying capacity (K) of the ODEs are calibrated using a simplified ODE model without an immune system and the tumor volumes of RAG1 KO mice. The parameters were chosen to minimize the mean squared error between actual and predicted tumor volumes. The proliferation rate (
) and the contact inhibition parameter (
) of tumor cells in the ABM were calibrated using the tumor volumes of RAG1 KO mice, and the Surrogate Modeling for Reconstructing Parameter Surfaces (SMoRe ParS) method developed by [
41]. SMoRe ParS employs an ODE surrogate model to estimate ABM parameter values from experimental data. The admissible parameter region for which
and
accurately capture the tumor growth data is shown in
Figure A1. From this region, we select baseline values for
and
so that we can focus on exploring the effects of other parameters related to the immune system and tumor antigenicity. In
Figure 2A, the blue line shows the mean volume of 25 virtual tumors with calibrated
and
. The range of simulated tumor volumes at each time point shows little variation, and the simulated trajectory closely matches the mean tumor volume of RAG1 KO mice, as shown in orange.
Fixing the calibrated
and
, we then varied ten other tumor-immune characteristics using Latin Hypercube Sampling in the range given in
Table 1 to construct a virtual cohort comprising 12,000 simulated TMEs. The initial conditions of the simulations are shown in
Table 2. Due to the stochastic nature of the ABM and computational limitations, we cannot vary all ABM parameters. We chose ten parameters that we believed would have the most impact on therapeutic outcomes based on the most sensitive parameters in the ODE model, which describes the same biological process, and our understanding of the spatial components of the ABM. In
Figure 2B, the blue line shows the median, interquartile range, and 95% simulated interval of tumor volumes up to Day 19. The orange lines show the mean and standard deviation of tumor volume of C57BL/6J mice on days when measurements were taken. The simulated trajectories lie reasonably close to the experimental data.
Based on the calculated density of CTLs within the tumor in C57BL/6J mice at the endpoint of Day 19, we estimated each ABM CTL represents 2175 actual CTL cells. This scale was calculated, and the range of the CTL recruitment rate in the ABM was chosen so that the range of simulated CTL densities on Day 19 in the virtual cohort matches the range observed experimentally, as illustrated in
Figure 2C. The green and orange lines show the observed minimum and maximum CTL density in C57BL/6J mice on Day 19. The grey dots show the endpoint CTL density of each virtual mouse, and the blue line shows the median CTL density for each integer interval of the CTL recruitment rate (e.g., 2–3, 19–20, etc.). The calibrated parameters are shown in
Table 3. All other ABM parameters are in
Table A1.
4. Discussion
We simulated tumor progression and the response to immune checkpoint blockade therapy in a virtual cohort using a three-dimensional, on-lattice ABM calibrated using in vivo data from bladder cancer studies in mice. Here, we present a comparison of an ODE model and an ABM for the same cancer immunotherapy: ICI for the PD-1/PD-L1 immune checkpoint. Our models reveal which tumor and immune characteristics affect the outcomes of checkpoint blockade therapy the most. While our previous work [
32] analyzed the ODE models thoroughly, this paper focuses on the capabilities of the ABM. In this way, we explore what biological insights both models can provide and what additional insights the ABM offers about the spatial complexity of the TME and its impact on therapeutic outcomes. Despite the enhanced modeling capabilities, the use of ABMs also presents challenges. Therefore, we will also discuss the pros and cons of the ODE model and the ABM for modeling tumor-immune dynamics.
The ODE model and ABM predict a wide range of therapeutic responses to immune checkpoint blockade therapy in a virtual cohort with similar tumor growth pre-treatment. Both models also identify crucial immune parameters linked to the range of outcomes. Our analysis of both models underscores the pivotal role of CTL recruitment rate (
) and maximum rate of antigen-mediated CTL proliferation (
) in tumor reduction or elimination. Since adoptive T-cell therapy can increase
and therapeutic cytokines like interleukin-2 (IL-2) can increase
, our results in
Figure 5A–D have implications on the effectiveness of combination therapy strategies. Our simulations suggest that combination therapy of anti-PD1 and adoptive T-cell transfer is effective in drastically reducing tumor size or eliminating tumors if the CTL recruitment rate can be enhanced to sufficiently high levels. Various combination therapy strategies involving anti-PD1 and tumor-infiltrating lymphocytes (TIL) or chimeric antigen receptor (CAR) T cells have shown synergistic effects in both preclinical studies and clinical trials [
44,
45]. Lifileucel, a TIL therapy, was recently approved for patients who received prior treatment with anti–PD1/PD-L1 antibodies [
45]. Our simulations suggest another possible way to achieve a drastic reduction of tumor volume or even tumor elimination with a smaller amount of drug: combining ICIs with both adoptive T-cell transfer and cytokine-directed therapy. In this way, a patient’s parameters can move from a baseline outcome of ICI monotherapy, where the tumor escapes with certainty, to a region in parameter space where tumor elimination is possible. In fact, IL-2 treatments are often administered with other forms of immunotherapy, such as Lifileucel. Furthermore, IL-2 therapy in combination with anti-PD1/PD-L1 was shown to be feasible and tolerable, although the clinical trials to show the effectiveness of this therapy are still underway [
46,
47].
Both models also show the importance of considering tumor antigenicity and multiple immune-cell kill mechanisms preferentially associated with HA or LA tumor cells. Our baseline assumption was that CTLs preferentially kill HA tumor cells via the fast mechanism and LA tumor cells via the slow mechanism. Effectively, we assumed that LA tumor cells are the harder-to-treat phenotype regarding antigenicity. Using virtual clones with different initial LA to total tumor cell ratios, we showed that the less LA-dominant the initial tumor is, the better the outcomes after immune checkpoint blockade. Moreover, the final tumor was always more LA-dominant than the initial tumor. These are both consequences of CTLs killing HA tumor cells faster than LA tumor cells. Higher numbers of LA tumor cells in the resulting tumor suggest that if ICI does not eliminate the tumor, it might become a “colder” tumor, thereby affecting the responses to subsequent treatments. The shift to LA-dominance aligns with well-documented observations of immune selection for lowly antigenic tumors [
48]. In the ABM, the antigenicity of the tumor cells not only determines how fast CTLs kill tumor cells once conjugation has occurred; it also greatly impacts the movement of CTLs before conjugation. CTLs gravitate towards regions with high ISF, and HA tumor cells secret higher ISF in our model. This key difference between HA and LA tumor cells underlies the impact of the CTL movement rate, conjugation rate, and LA-ISF factors on treatment outcomes.
The ABM enhances our understanding of the TME by incorporating spatial characteristics that ODEs cannot capture. This allows for more nuanced insights, revealing complexities that might be overlooked when immune parameters, initial tumor composition, and the temporal evolution of cellular populations appear similar. In both models, we observed the importance of CTL recruitment rate (
) and max antigen-stimulated CTL proliferation rate (
) to tumor elimination after immune checkpoint blockade. This might seem intuitive as higher
and
results in more active CTLs in the TME, and thus, they eliminate more tumor cells. However, we chose 603 virtual tumors from ABM simulations to show that, when considering intratumoral spatial heterogeneity, tumors with similar
and
values and similar temporal trajectories of CTLs in the TME can experience drastically different fates after checkpoint blockade therapy (
Figure 6 and
Figure 7). In the ODE model formulation, CTLs indiscriminately target all HA or all LA. By contrast, in the ABM, immune attacks are contingent on CTLs moving toward tumor cells and successfully conjugating with them. Therefore, the movement rate of CTLs (
m) and the conjugation rate of CTLs with tumor cells (
) prove to be crucial in determining how fast CTLs colocalize to, attack, and clear tumor cells. Virtual TMEs with high
m and
are more likely to get tumor elimination after checkpoint blockade.
Translational data are emerging on the critical nature of spatial relationships in the immune tumor microenvironment. A multitude of factors, such as gradients of chemokines and physical features of the microenvironment, have been shown to affect T cell movement [
49]. In a melanoma mouse study, adoptive T-cell therapy successfully controlled tumor growth in some cases but failed in others. The T-cell infiltration and motility were higher in responders relative to non-responders, as evidenced by increased speed and distance traveled of T cells [
50]. An in vitro study of melanoma showed that varied ICI responses were not merely due to differences in tumor structure or proportion of cell types. Physical proximity and contact frequency between CTLs and tumor cells also significantly differed between responders and nonresponders of ICIs [
51]. Among many ongoing efforts to develop therapeutics to enhance T-cell motility and infiltration, tebentafusp, a bispecific protein consisting of an affinity-enhanced T-cell receptor fused to an anti-CD3 effector that can redirect T cells to target glycoprotein 100–positive cells [
52], was FDA-approved in 2022.
With proper formulation, both ABMs and ODE models can accurately reflect biological processes. Still, they have a few fundamental differences, which lead to their respective pros and cons from the modeling perspective. ODEs model the population-level temporal dynamics of each type of cell or drug molecule, whereas ABMs model each cell as an autonomous agent. At a given time point, all cells or molecules of a single type in the ODE undergo the same changes uniformly, whereas agents in the ABM experience different events based on their location in space or what other agents surround them. The ABM captures the phenotypical heterogeneity of the three-dimensional tumor in space and the spatial activities of CTLs in the TME. This approach allows us to obtain the spatial distribution of CTLs with respect to each tumor cell and the frequency with which each CTL attacks and clears tumor cells. These individual cell-level insights explain the diverging outcomes of immune checkpoint blockade in virtual tumors despite a similar total number of CTLs in the TME. Such details cannot be obtained in continuous differential equation-based models. Thus, the ABM is undoubtedly more flexible in modeling intratumoral differences and more closely reflects complexities seen in vivo. Moreoever, the discrete and stochastic nature of ABMs, contrasted with the continuous and deterministic nature of ODEs, might have caused the ABM to wander away from locally stable equilibrium, leading to what we observed in
Figure 3C,D. Tumor growths reach equilibrium faster in the ABM than in the ODE model, leading to the lack of intermediate-sized tumors on Day 19 in the ABM. The enhanced granularity and versatility of ABMs come at the cost of longer computational time and increased difficulty in parameterizing and analyzing the model. Because the ABM updates each cell individually at each time step, simulations slow down significantly when the number of tumor cells increases exponentially. Thus, simulating tumor and immune dynamics at a realistic scale is computationally prohibitive. ABMs generally have many more parameters than the ODE model, making parametrization of the model challenging. In the ODE, we used sensitivity analysis to determine which parameters impact the tumor outcome most and focused our calibration and analytical efforts on those. Sensitivity analysis of ABM parameters, though possible [
53,
54], is no trivial task. Future avenues of exploration include using machine learning to overcome the shortcomings of ABMs. In our upcoming work, we plan to combine this ABM with machine learning algorithms to predict the tumor-immune landscape after immune checkpoint blockade. This can make simulating larger virtual cohorts or a larger number of cells more feasible. With future developments in efficient simulations and global sensitivity analysis of ABMs, we are also interested in exploring more regions of the parameters space and comparing their impact on the TME with what we observe in this paper.