Quantifying Intratumoral Heterogeneity and Immunoarchitecture Generated In-Silico by a Spatial Quantitative Systems Pharmacology Model

Simple Summary We introduced a novel approach to quantitatively validate the performance of a hybrid spatio-temporal method called spatial quantitative systems pharmacology (spQSP). This platform is composed of a compartmental QSP model describing tumor growth dynamics, anti-tumor immune response and immune checkpoint therapy in a whole-patient and a spatial agent-based model, describing the tumor to simulate the effect of anti-PD-1 therapy (an immune checkpoint inhibitor) on simulated intratumoral heterogeneity. Four spatial metrics adopted from computational digital pathology, along with the ratio of cancer cells to immune cells, were used to categorize the tumor microenvironment as “cold”, “mixed” and “compartmentalized” patterns, which were related to the efficacy of the treatment. This study compared the intratumoral heterogeneity description capability of the metrics to facilitate future comprehensive and tangible research on specific cancer types, different therapeutics as single agents or combination therapies, and immunopathological multiplexed samples. Having a better quantitative understanding of intratumoral heterogeneity using numerical simulations can help design more effective treatments. Abstract Spatial heterogeneity is a hallmark of cancer. Tumor heterogeneity can vary with time and location. The tumor microenvironment (TME) encompasses various cell types and their interactions that impart response to therapies. Therefore, a quantitative evaluation of tumor heterogeneity is crucial for the development of effective treatments. Different approaches, such as multiregional sequencing, spatial transcriptomics, analysis of autopsy samples, and longitudinal analysis of biopsy samples, can be used to analyze the intratumoral heterogeneity (ITH) and temporal evolution and to reveal the mechanisms of therapeutic response. However, because of the limitations of these data and the uncertainty associated with the time points of sample collection, having a complete understanding of intratumoral heterogeneity role is challenging. Here, we used a hybrid model that integrates a whole-patient compartmental quantitative-systems-pharmacology (QSP) model with a spatial agent-based model (ABM) describing the TME; we applied four spatial metrics to quantify model-simulated intratumoral heterogeneity and classified the TME immunoarchitecture for representative cases of effective and ineffective anti-PD-1 therapy. The four metrics, adopted from computational digital pathology, included mixing score, average neighbor frequency, Shannon’s entropy and area under the curve (AUC) of the G-cross function. A fifth non-spatial metric was used to supplement the analysis, which was the ratio of the number of cancer cells to immune cells. These metrics were utilized to classify the TME as “cold”, “compartmentalized” and “mixed”, which were related to treatment efficacy. The trends in these metrics for effective and ineffective treatments are in qualitative agreement with the clinical literature, indicating that compartmentalized immunoarchitecture is likely to result in more efficacious treatment outcomes.

[1]  Joshua A. Bull,et al.  Quantification of spatial and phenotypic heterogeneity in an agent-based model of tumour-macrophage interactions , 2023, PLoS Comput. Biol..

[2]  M. Robertson-Tessi,et al.  Agent-based methods facilitate integrative science in cancer. , 2022, Trends in cell biology.

[3]  Aman P. Singh,et al.  From Cold to Hot: Changing Perceptions and Future Opportunities for Quantitative Systems Pharmacology Modeling in Cancer Immunotherapy , 2022, Clinical pharmacology and therapeutics.

[4]  E. Jaffee,et al.  Quantitative Spatial Profiling of Immune Populations in Pancreatic Ductal Adenocarcinoma Reveals Tumor Microenvironment Heterogeneity and Prognostic Biomarkers , 2022, Cancer research.

[5]  S. De,et al.  Inference on spatial heterogeneity in tumor microenvironment using spatial transcriptomics data , 2022, Computational and systems oncology.

[6]  Ming Y. Lu,et al.  Pan-cancer integrative histology-genomic analysis via multimodal deep learning. , 2022, Cancer cell.

[7]  K. Polyak,et al.  Untangling the web of intratumour heterogeneity , 2022, Nature Cell Biology.

[8]  O. Grimm,et al.  Integrating digital pathology and mathematical modelling to predict spatial biomarker dynamics in cancer immunotherapy , 2022, npj Digital Medicine.

[9]  A. Popel,et al.  Simulations of tumor growth and response to immunotherapy by coupling a spatial agent-based model with a whole-patient quantitative systems pharmacology model , 2022, PLoS Comput. Biol..

[10]  A. Popel,et al.  Dynamics of tumor-associated macrophages in a quantitative systems pharmacology model of immunotherapy in triple-negative breast cancer , 2022, iScience.

[11]  A. Madabhushi,et al.  The state of the art for artificial intelligence in lung digital pathology , 2022, The Journal of pathology.

[12]  A. Popel,et al.  Multi-Scale Spatial Analysis of the Tumor Microenvironment Reveals Features of Cabozantinib and Nivolumab Efficacy in Hepatocellular Carcinoma , 2022, Frontiers in Immunology.

[13]  E. Nakakura,et al.  Discovering dominant tumor immune archetypes in a pan-cancer census , 2021, Cell.

[14]  A. Popel,et al.  825 High-dimensional image cytometry reveals spatially organized tumor-immune microenvironment in hepatocellular carcinoma , 2021, Journal for ImmunoTherapy of Cancer.

[15]  V. Muzykantov,et al.  A numerical study on drug delivery via multiscale synergy of cellular hitchhiking onto red blood cells. , 2021, Nanoscale.

[16]  A. Popel,et al.  A Spatial Quantitative Systems Pharmacology Platform spQSP-IO for Simulations of Tumor–Immune Interactions and Effects of Checkpoint Inhibitor Immunotherapy , 2021, Cancers.

[17]  A. Popel,et al.  Integrating single cell sequencing with a spatial quantitative systems pharmacology model spQSP for personalized prediction of triple-negative breast cancer immunotherapy response , 2021, Immunoinformatics.

[18]  M. Nikfar,et al.  Small molecule therapeutics to destabilize the ACE2-RBD complex: A molecular dynamics study , 2021, Biophysical Journal.

[19]  Z. Yakhini,et al.  Assessing heterogeneity in spatial data using the HTA index with applications to spatial transcriptomics and imaging , 2021, bioRxiv.

[20]  Nathaniel J. Merrill,et al.  History and Future Perspectives on the Discipline of Quantitative Systems Pharmacology Modeling and Its Applications , 2021, Frontiers in Physiology.

[21]  S. Handelman,et al.  Standing Variations Modeling Captures Inter-Individual Heterogeneity in a Deterministic Model of Prostate Cancer Response to Combination Therapy , 2021, bioRxiv.

[22]  A. Popel,et al.  Quantitative systems pharmacology model predictions for efficacy of atezolizumab and nab-paclitaxel in triple-negative breast cancer , 2021, Journal for ImmunoTherapy of Cancer.

[23]  Stacey D. Finley,et al.  Multi-scale modeling of macrophage—T cell interactions within the tumor microenvironment , 2020, PLoS Comput. Biol..

[24]  A. Szalay,et al.  Digital Pathology Analysis Quantifies Spatial Heterogeneity of CD3, CD4, CD8, CD20, and FoxP3 Immune Markers in Triple-Negative Breast Cancer , 2020, Frontiers in Physiology.

[25]  A. Fletcher,et al.  Seven challenges in the multiscale modeling of multicellular tissues. , 2020, WIREs mechanisms of disease.

[26]  Z. Yakhini,et al.  Spatial transcriptomics inferred from pathology whole-slide images links tumor heterogeneity to survival in breast and lung cancer , 2020, Scientific Reports.

[27]  K. Polyak,et al.  Intratumor Heterogeneity: The Rosetta Stone of Therapy Resistance. , 2020, Cancer cell.

[28]  A. Popel Immunoactivating the tumor microenvironment enhances immunotherapy as predicted by integrative computational model , 2020, Proceedings of the National Academy of Sciences.

[29]  J. Taube,et al.  Neoadjuvant checkpoint blockade for cancer immunotherapy , 2020, Science.

[30]  Leping Li,et al.  Putative biomarkers for predicting tumor sample purity based on gene expression data , 2019, BMC Genomics.

[31]  B. Wang,et al.  A Computational Model of Neoadjuvant PD-1 Inhibition in Non-Small Cell Lung Cancer , 2019, The AAPS Journal.

[32]  Justin C. Earp,et al.  Translational Quantitative Systems Pharmacology in Drug Development: from Current Landscape to Good Practices , 2019, The AAPS Journal.

[33]  A. Popel,et al.  Tumor Ensemble-Based Modeling and Visualization of Emergent Angiogenic Heterogeneity in Breast Cancer , 2019, Scientific Reports.

[34]  Martial Guillaud,et al.  Hyperspectral cell sociology reveals spatial tumor-immune cell interactions associated with lung cancer recurrence , 2019, Journal of Immunotherapy for Cancer.

[35]  Bing Wang,et al.  Quantitative Characterization of CD8+ T Cell Clustering and Spatial Heterogeneity in Solid Tumors , 2019, Front. Oncol..

[36]  Horst K. Hahn,et al.  Data-Driven Discovery of Immune Contexture Biomarkers , 2018, Front. Oncol..

[37]  N. Razavian,et al.  Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning , 2018, Nature Medicine.

[38]  A. Madabhushi,et al.  Spatial Architecture and Arrangement of Tumor-Infiltrating Lymphocytes for Predicting Likelihood of Recurrence in Early-Stage Non–Small Cell Lung Cancer , 2018, Clinical Cancer Research.

[39]  Jakob Nikolas Kather,et al.  Topography of cancer-associated immune cells in human solid tumors , 2018, eLife.

[40]  Sean C. Bendall,et al.  A Structured Tumor-Immune Microenvironment in Triple Negative Breast Cancer Revealed by Multiplexed Ion Beam Imaging , 2018, Cell.

[41]  Boris Sepesi,et al.  Effect of neoadjuvant chemotherapy on the immune microenvironment in non–small cell lung carcinomas as determined by multiplex immunofluorescence and image analysis approaches , 2018, Journal of Immunotherapy for Cancer.

[42]  K. Urbanek,et al.  Blood and lymphatic vessels contribute to the impact of the immune microenvironment on clinical outcome in non-small-cell lung cancer† , 2018, European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery.

[43]  Rajarsi R. Gupta,et al.  Spatial Organization and Molecular Correlation of Tumor-Infiltrating Lymphocytes Using Deep Learning on Pathology Images. , 2018, Cell reports.

[44]  Souptik Barua,et al.  Spatial interaction of tumor cells and regulatory T cells correlates with survival in non-small cell lung cancer. , 2018, Lung cancer.

[45]  A. Shaw,et al.  Tumour heterogeneity and resistance to cancer therapies , 2018, Nature Reviews Clinical Oncology.

[46]  A. Antabak,et al.  Tumor angiogenesis as an important prognostic factor in advanced non-small cell lung cancer (Stage IIIA) , 2017, Oncology letters.

[47]  A. Radunskaya,et al.  Mathematical Modeling of Tumor Immune Interactions: A Closer Look at the Role of a PD-L1 Inhibitor in Cancer Immunotherapy , 2018 .

[48]  A. Popel,et al.  A computational multiscale agent-based model for simulating spatio-temporal tumour immune response to PD1 and PDL1 inhibition , 2017, Journal of The Royal Society Interface.

[49]  Aleksander S. Popel,et al.  An agent-based model of triple-negative breast cancer: the interplay between chemokine receptor CCR5 expression, cancer stem cells, and hypoxia , 2017, BMC Systems Biology.

[50]  Samuel H. Friedman,et al.  BioFVM: an efficient, parallelized diffusive transport solver for 3-D biological simulations , 2015, Bioinform..

[51]  Aleksander S Popel,et al.  An agent-based model of cancer stem cell initiated avascular tumour growth and metastasis: the effect of seeding frequency and location , 2014, Journal of The Royal Society Interface.

[52]  D. Rimm,et al.  Multiplexed Quantitative Analysis of CD3, CD8, and CD20 Predicts Response to Neoadjuvant Chemotherapy in Breast Cancer , 2014, Clinical Cancer Research.

[53]  Aleksander S Popel,et al.  A bioimage informatics based reconstruction of breast tumor microvasculature with computational blood flow predictions. , 2014, Microvascular research.

[54]  C. C. Law,et al.  ParaView: An End-User Tool for Large-Data Visualization , 2005, The Visualization Handbook.