Abstract

Significant scientific advances in biomedical research have expanded our knowledge of the molecular basis of carcinogenesis, mechanisms of cancer growth, and the importance of the cancer immunity cycle. However, despite scientific advances in the understanding of cancer biology, the success rate of oncology drug development remains the lowest among all therapeutic areas. In this review, some of the key translational drug development objectives in oncology will be outlined. The literature evidence of how mathematical modeling could be used to build a unifying framework to answer these questions will be summarized with recommendations on the strategies for building such a mathematical framework to facilitate the prediction of clinical efficacy and toxicity of investigational antineoplastic agents. Together, the literature evidence suggests that a rigorous and unifying preclinical to clinical translational framework based on mathematical models is extremely valuable for making go/no-go decisions in preclinical development, and for planning early clinical studies.

Highlights

  • There are a number of mathematical approaches to translate the preclinically observed antitumor activity into clinical efficacy

  • These approaches can generally be categorized into static algebraic approaches and dynamic, differential equations-based approaches

  • A model-based development paradigm will result in a rational and more efficient oncology drug development process

Read more

Summary

Slow tumor Fast tumor

Study durations compared with the T/C ratio and TGI [21]. Thirdly, compared with T/C or TGI, GRI is less influenced by the intrinsic growth rate of the xenograft tumor. GRI has a much more dynamic range compared with TGI in fast-growing tumors. For xenograft tumors with faster growth rates, GRI has a much more dynamic range compared with TGI, which saturates at around 100% in fast-growing tumors. There is often a delay between drug administration and tumor shrinkage (i.e., it takes a while for the drug to kill the tumor cells) which cannot be accounted for by static approaches. To overcome these limitations, a number of differential equation-based dynamic approaches have been developed to describe the tumor growth and exposure–response relationship of antineoplastic agents.

Growth rates
Phase nonspecific models
Predicted clinical therapeutic margin
Antitumor activity in xenograft models correlates with clinical response
Other types of toxicity
Conclusion & future perspective
Executive summary
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