Abstract

The ability to rapidly assess the efficacy of therapeutic strategies for incurable bone metastatic prostate cancer is an urgent need. Pre-clinical in vivo models are limited in their ability to define the temporal effects of therapies on simultaneous multicellular interactions in the cancer-bone microenvironment. Integrating biological and computational modeling approaches can overcome this limitation. Here, we generated a biologically driven discrete hybrid cellular automaton (HCA) model of bone metastatic prostate cancer to identify the optimal therapeutic window for putative targeted therapies. As proof of principle, we focused on TGFβ because of its known pleiotropic cellular effects. HCA simulations predict an optimal effect for TGFβ inhibition in a pre-metastatic setting with quantitative outputs indicating a significant impact on prostate cancer cell viability, osteoclast formation and osteoblast differentiation. In silico predictions were validated in vivo with models of bone metastatic prostate cancer (PAIII and C4-2B). Analysis of human bone metastatic prostate cancer specimens reveals heterogeneous cancer cell use of TGFβ. Patient specific information was seeded into the HCA model to predict the effect of TGFβ inhibitor treatment on disease evolution. Collectively, we demonstrate how an integrated computational/biological approach can rapidly optimize the efficacy of potential targeted therapies on bone metastatic prostate cancer.

Highlights

  • Automata (HCA), are better suited to test hypotheses using a mechanistic “bottom-up” approach to provide unbiased predictions[16]

  • To obtain a realistic readout of the level of TGFβinhibition that could be achievable in vivo, we treated mice with a TGFβneutralizing antibody (1D11) at a dose previously used in the literature (10 mg/kg) and consistent with clinical trials performed for a humanized version of the 1D11 TGFβneutralizing antibody, fresolimumab/GC100822,29,30

  • We observed that the TGFβneutralizing antibody significantly reduced circulating TGFβserum levels by up to 80% compared to IgG control treated animals (Supplementary Fig. S1)

Read more

Summary

Introduction

Automata (HCA), are better suited to test hypotheses using a mechanistic “bottom-up” approach to provide unbiased predictions[16] These models work by parameterizing the properties of cells (or agents) with regards to proliferation, apoptosis, secretion of factors, genetic mutations or even metabolism[17]. TGFβinhibitors such as neutralizing antibodies are currently undergoing clinical trial[22] Their application for the treatment of osteogenic bone metastatic prostate cancer has not been explored far due to the pleiotropic and often opposing effects TGFβcan have on cancer and bone cell behavior[5,23,24,25]. We demonstrate how an integrated computational/biological modeling approach can be used to optimize therapy efficacy for the treatment of bone metastatic prostate cancer

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call