Skip to main content

Emergence of Anti-Cancer Drug Resistance: Exploring the Importance of the Microenvironmental Niche via a Spatial Model

  • Conference paper

Part of the book series: The IMA Volumes in Mathematics and its Applications ((IMA,volume 158))

Abstract

Practically, all chemotherapeutic agents lead to drug resistance. Clinically, it is a challenge to determine whether resistance arises prior to, or as a result of, cancer therapy. Further, a number of different intracellular and microenvironmental factors have been correlated with the emergence of drug resistance. With the goal of better understanding drug resistance and its connection with the tumor microenvironment, we have developed a hybrid discrete-continuous mathematical model. In this model, cancer cells described through a particle-spring approach respond to dynamically changing oxygen and DNA damaging drug concentrations described through partial differential equations. We thoroughly explored the behavior of our self-calibrated model under the following common conditions: a fixed layout of the vasculature, an identical initial configuration of cancer cells, the same mechanism of drug action, and one mechanism of cellular response to the drug. We considered one set of simulations in which drug resistance existed prior to the start of treatment, and another set in which drug resistance is acquired in response to treatment. This allows us to compare how both kinds of resistance influence the spatial and temporal dynamics of the developing tumor, and its clonal diversity. We show that both pre-existing and acquired resistance can give rise to three biologically distinct parameter regimes: successful tumor eradication, reduced effectiveness of drug during the course of treatment (resistance), and complete treatment failure. When a drug resistant tumor population forms from cells that acquire resistance, we find that the spatial component of our model (the microenvironment) has a significant impact on the transient and long-term tumor behavior. On the other hand, when a resistant tumor population forms from pre-existing resistant cells, the microenvironment only has a minimal transient impact on treatment response. Finally, we present evidence that the microenvironmental niches of low drug/sufficient oxygen and low drug/low oxygen play an important role in tumor cell survival and tumor expansion. This may play role in designing new therapeutic agents or new drug combination schedules.

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

References

  1. J.J. Kim, I.F. Tannock (2005) Repopulation of cancer cells during therapy: an important cause of treatment failure, Nat. Rev. Cancer, 5:516–525.

    Article  Google Scholar 

  2. M. Dean, T. Fojo, S Bates (2005) Tumour stem cells and drug resistance, Nat. Rev. Cancer, 5(4):275–284.

    Article  Google Scholar 

  3. B. Baguley (2010) Multiple drug resistance mechanisms in cancer., Mol. Biotechnol. 46(3):308–316.

    Google Scholar 

  4. H. Zahreddine, K.L B. Borden (2013) Mechanisms and insights into drug resistance in cancer, Front. Pharmacol. 4:e28.

    Article  Google Scholar 

  5. C. Bock, T. Lengauer (2012) Managing drug resistance in cancers: lessons from HIV therapy, Nat. Rev. Cancer, 12:494–501.

    Article  Google Scholar 

  6. G. Lambert, L. Estévez-Salmeron, S. Oh, D. Liao, B.M. Emerson, T.D. Tlsty, R.H. Austin (2011) An analogy between the evolution of drug resistance in bacterial communities and malignant tissues. Nat. Rev. Cancer, 11:375–382.

    Article  Google Scholar 

  7. O. Trédan, C.M. Galmarini, K. Patel, I.F. Tannock (2007) Drug resistance and the solid tumor microenvironment., J Natl Cancer Inst, 99(19):1441–1454.

    Article  Google Scholar 

  8. A. Wu, K. Loutherback, G. Lambert, L. Estévez-Salmeron, T.D. Tlsty, R.H. Austin, J.C. Sturma (2013) Cell motility and drug gradients in the emergence of resistance to chemotherapy, Proc. Natl. Acad. Sci. 110(40):16103–16108.

    Article  Google Scholar 

  9. S. Rottenberg, A.O. H. Nygren, M. Pajic, F.W. B. van Leeuwen, I. van der Heijden, K. van de Wetering, X. Liu, K.E. de Visser, K.G. Gilhuijs, O. van Tellingen, J.P. Schouten, J. Jonkers, P. Borst (2007) Selective induction of chemotherapy resistance of mammary tumors in a conditional mouse model for hereditary breast cancer, Proc. Natl. Acad. Sci. 104(29):12117–12122.

    Article  Google Scholar 

  10. A.L. Correia, M.J. Bissell (2012) The tumor microenvironment is a dominant force in multidrug resistance, Drug Resist. Update, 15:39–49.

    Google Scholar 

  11. H. Lage. An overview of cancer multidrug resistance: a still unsolved problem (2008) Cellular and Mol. Life Sci. 65:3145–3167.

    Google Scholar 

  12. K. Cheung-Ong, G. Giaever, C. Nislow (2013) DNA-Damaging Agents in Cancer Chemotherapy: Serendipity and Chemical Biology, Cell Chem. Biol. 20:648–659.

    Google Scholar 

  13. D. Woods, J.J. Turchi (2013) Chemotherapy induced DNA damage response Convergence of drugs and pathways, Cancer Biol. Ther. 14(5):379–389.

    Google Scholar 

  14. M.B. Meads, R.A. Gatenby, W.S. Dalton (2009) Environment-mediated drug resistance: a major contributor to minimal residual disease., Nat. Rev. Cancer, 9:665–674.

    Google Scholar 

  15. E.S. Nakasone, H.A. Askautrud, T. Kees, J.H Park, V. Plaks, A.J. Ewald, M. Fein, M.G. Rasch, Y.X Tan, J. Qiu, J. Park, P. Sinha, M.J. Bissell, E. Frengen, Z. Werb, M. Egeblad (2012) Imaging Tumor-Stroma Interactions during Chemotherapy Reveals Contributions of the Microenvironment to Resistance., Cancer Cell 21:488–503.

    Article  Google Scholar 

  16. D.W. McMillin, J.M. Negri, C.S. Mitsiades (2013) The role of tumour-stromal interactions in modifying drug response: challenges and opportunities, Nat. Rev. Drug Discov, 12:217–228.

    Article  Google Scholar 

  17. P.P. Provenzano, S.R. Hingorani (2013) Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer, 108:1–8.

    Article  Google Scholar 

  18. P. Karran (2001) Mechanisms of tolerance to DNA damaging therapeutic drugs, Carcinogenesis, 22(12):1931–1937.

    Article  Google Scholar 

  19. M. Sawicka, M. Kalinowska, J. Skierski, W. Lewandowski (2004) A review of selected anti-tumour therapeutic agents and reasons for multidrug resistance occurrence., Pharm. Pharmacol. 56:1067–1081.

    Article  Google Scholar 

  20. F.A. Meineke, C.S. Potten, M. Loeffler (2001) Cell migration and organization in the intestinal crypt using a lattice-free model. Cell Prolif., 34:253–266.

    Article  Google Scholar 

  21. H.M. Byrne (2010) Dissecting cancer through mathematics: from the cell to the animal model, Nat. Rev. Cancer, 10(3):221–230.

    Article  Google Scholar 

  22. N.L. Komarova, D. Wodarz (2005) Drug resistance in cancer: Principles of emergence and prevention, Proc. Natl. Acad. Sci. 102(27):9714–9719.

    Article  Google Scholar 

  23. O. Lavi, M.M. Gottesman, D. Levy (2012) The dynamics of drug resistance: A mathematical perspective. Drug Resist Update, 15:90–97.

    Article  Google Scholar 

  24. T. Brocato, P. Dogra, E. J. Koay, A. Day, Y-L. Chuang, Z. Wang, V. Cristini (2014) Understanding drug resistance in breast cancer with mathematical oncology. Curr. Breast Cancer Rep, 6(2):110–120.

    Article  Google Scholar 

  25. J. Foo, F. Michor (2014) Evolution of acquired resistance to anti-cancer therapy. J Theor. Biol. 355:10–20.

    Article  Google Scholar 

  26. N.L. Komarova, D. Wodarz (2007) Stochastic modeling of cellular colonies with quiescence: An application to drug resistance in cancer. Theor. Popul. Biol. 72(4):523–538.

    Article  MATH  Google Scholar 

  27. J.J. Cunningham, R.A. Gatenby, J.S. Brown (2011) Evolutionary dynamics in cancer therapy. Mol. Pharm. 8(6):2094–2100.

    Article  Google Scholar 

  28. S.M. Mumenthaler, J. Foo, K. Leder, N.C. Choi, D.B. Agus, W. Pao, P. Mallick, F. Michor (2011) Evolutionary modeling of combination treatment strategies to overcome resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Mol. Pharm. 8(6):2069–2079.

    Article  Google Scholar 

  29. J. Foo, K. Leder, S.M. Mumenthaler (2013) Cancer as a moving target: understanding the composition and rebound growth kinetics of recurrent tumors. Evol. Appl., 6:54–69.

    Article  Google Scholar 

  30. I. Bozic, J.G. Reiter, B. Allen, T. Antal, K. Chatterjee, P. Shah, Y. S. Moon, A. Yaqubie, N. Kelly, D.T. Le, E.J. Lipson, P.B. Chapman, L.A. Diaz, B. Vogelstein, M.A. Nowak (2013) Evolutionary dynamics of cancer in response to targeted combination therapy. eLife, 2:1–15.

    Article  Google Scholar 

  31. P.A. Orlando, R.A. Gatenby, J.S. Brown (2012) Cancer treatment as a game: integrating evolutionary game theory into the optimal control of chemotherapy. Phys. Biol. 9(6):065007.

    Article  Google Scholar 

  32. A.O. Pisco, A. Brock, J. Ahou, A. Moor, M. Mojtahedi, D. Jackson, S. Huang (2013) Non-Darwinian dynamics in therapy-induced cancer drug resistance, Nat. Commun. 4:2467.

    Article  Google Scholar 

  33. M.M. Hadjiandreou, G.D. Mitsis (2013) Mathematical modeling of tumor growth, drug-resistance, toxicity, and optimal therapy design, IEEE Trans. Biomed. Eng. 61(2):415–425.

    Article  Google Scholar 

  34. T.L. Jackson, H.M. Byrne (2000) A mathematical model to study the effects of drug resistance and vasculature on the response of solid tumors to chemotherapy, Math. Biosci. 164:17–38.

    Article  MathSciNet  MATH  Google Scholar 

  35. J.J. Lee, J. Huang, C.G. England, L.R. McNally, H.B. Frieboes (2013) Predictive modeling of in vivo response to gemcitabine in pancreatic cancer, PLoS Comput. Biol. 9(9):e1003231.

    Google Scholar 

  36. A.S. Silva, R.A. Gatenby (2010) A theoretical quantitative model for evolution of cancer chemotherapy resistance, Biol. Direct. 5(1):25.

    Article  Google Scholar 

  37. A. Lorz, T. Lorenzi, M.E. Hochberg, J. Clairambault, B. Perthame (2013) Populational adaptive evolution, chemotherapeutic resistance and multiple anti-cancer therapies, ESAIM: Math. Model. Num. Anal. 47:377–399.

    Article  MathSciNet  MATH  Google Scholar 

  38. A. Lorz, T. Lorenzi, J. Clairambault, A. Escargueil, B. Perthame (2015) Modeling effects of space structure and combination therapies on phenotypic heterogeneity and drug resistance in solid tumors, Bull. Math. Biol. 77(1):1–2

    Article  MathSciNet  Google Scholar 

  39. O. Lavi, J.M. Greene, D. Levy, M.M. Gottesman (2013) The role of cell density and intratumoral heterogeneity in multidrug resistance. Canc. Res. 73(24):7168–7175.

    Article  Google Scholar 

  40. J. Greene, O. Lavi, M.M. Gottesman, D. Levy (2014) The impact of cell density and mutations in a model of multidrug resistance in solid tumors, Bull. Math. Biol. 76(3):627–653.

    Article  MathSciNet  Google Scholar 

  41. G.G. Powathil, M.A. Chaplain, M. Swat (in press) Investigating the development of chemotherapeutic drug resistance in cancer: a multiscale computational study, IET Sys. Biol.

    Google Scholar 

Download references

Acknowledgements

We thank Drs. Ami Radunskaya and Trachette Jackson for organizing the WhAM! Research Collaboration Workshop for Women in Applied Mathematics, Dynamical Systems and Applications to Biology and Medicine, that allowed us to initiate this research project, and the Institute for Mathematics and Its Applications (IMA) for a generous support during the WhAM! workshop and reunion meetings. We also acknowledge the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG) for the travel grant (CE 243/1-1).

KAN was supported by T32 CA13084005 and an American Cancer Society postdoctoral fellowship. JPV would like to thank the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG) for providing a travel grant (CE 243/1-1) to facilitate international cooperation. AV was supported by NSF Graduate Research Fellowship (GRFP) under Grant No. DGE0228243. KAR was supported in part by the NIH grants U54-CA113007 and U54-CA-143970, and the Institutional Research Grant number 93-032-16 from the American Cancer Society.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Katarzyna A. Rejniak .

Editor information

Editors and Affiliations

1 Electronic Supplementary Material

Appendix

Appendix

Before testing various mechanisms of tumor resistance, our model has been calibrated to 1) achieve a stable gradient of oxygen when no cancer cells are present, as would be the case in healthy tissue; 2) generate a tumor cluster that completely fills the available space when no drug is applied as would take place in non-treated tumors; this will result in another stable gradient of oxygen with hypoxic areas located far from the vasculature; and 3) completely eliminate the tumor during the treatment when the cells do not acquire resistance. These three self-calibration steps are discussed in this section.

First, the influence of oxygen and drug are normalized so that S ξ  = S γ  = 1. Then we determine the oxygen diffusion coefficient \(\mathcal{D}_{\xi }\) and oxygen boundary conditions that lead to a (numerically) stable gradient of oxygen when no cancer cells are present; that is with no cellular uptake. Several boundary conditions were considered, however, the best results in terms of irregular gradient stabilization and the extent of hypoxic areas were achieved for the sink-like conditions with \(\varpi =\) 0.45 (see Section 2.1). The resulting stable oxygen gradient is shown in Fig. 9a and the relative 2-norm error between two oxygen concentrations generated in two consecutive time steps is shown in Fig. 9c. The obtained oxygen gradient served as an initial condition for the reaction-diffusion equation for oxygen.

Next, a tumor cell oxygen uptake rate p ξ and a hypoxia threshold Thr hypo were selected to allow for tumor growth with a small subpopulation of hypoxic cells, and for generation of a (numerically) stable gradient of oxygen when the tumor reaches its stable configuration. This population of tumor cells, including the hypoxic cell fraction and tumor clonal composition, serves as a control case (with no treatment) shown in Fig. 1b. For the stable tumor population the (numerically) stable oxygen gradient is shown in Fig. 9b, and the relative 2-norm error of the oxygen changes over 25, 000 iterations is shown in Fig. 9d.

Fig. 9
figure 9

Oxygen distribution and its stabilization error curves. (a,b) Numerically stable gradients of oxygen in a domain with no cancer cells (a), and in a domain where non-treated cancer cells uptake oxygen (b). The greyscale contours indicate oxygen distribution levels. (c,d) Relative 2-norm error of oxygen changes \(\|\eta (\mathbf{x},t +\varDelta t) -\eta (\mathbf{x},t)\|_{2}\) calculated for 25000 iterations showing its numerical stability for cases (a) and (b), respectively.

Finally, the drug diffusion coefficient \(\mathcal{D}_{\gamma }\), drug uptake rate p γ , and death threshold Thr death were determined so that the cluster of tumor cells with no resistance is eradicated. This ensures that for the chosen drug parameters, the drug is effective when there are no resistant tumor cells. This case is discussed in Section 3.1. All parameters determined by the procedure described here are listed in Table 2.

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer Science+Business Media, LLC

About this paper

Cite this paper

Gevertz, J.L., Aminzare, Z., Norton, KA., Pérez-Velázquez, J., Volkening, A., Rejniak, K.A. (2015). Emergence of Anti-Cancer Drug Resistance: Exploring the Importance of the Microenvironmental Niche via a Spatial Model. In: Jackson, T., Radunskaya, A. (eds) Applications of Dynamical Systems in Biology and Medicine. The IMA Volumes in Mathematics and its Applications, vol 158. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-2782-1_1

Download citation

Publish with us

Policies and ethics