Skip to main content

Cancer Chemotherapy for Heterogeneous Tumor Cell Populations and Drug Resistance

  • Chapter
Optimal Control for Mathematical Models of Cancer Therapies

Part of the book series: Interdisciplinary Applied Mathematics ((IAM,volume 42))

  • 1756 Accesses

Abstract

The results of the previous chapter are consistent with the classical MTD paradigm in medicine: give as much of the drug as possible immediately. This makes perfect sense in many situations: cancer is a widely symptomless disease which, once finally detected, often is in an advanced stage where immediate action is required. Then the aim simply is to be as toxic as possible to the cancerous cells. However, this presumes that cells can be killed, i.e., that the tumor population consists of chemotherapeutically sensitive cells. Malignant cancer cell populations on the other hand are often highly genetically unstable and coupled with fast proliferation rates; this leads to a great variety in the structure of the cells within one tumor—the number of genetic errors present within one cancer cell can lie in the thousands [220]. Consequently, many tumors consist of heterogeneous agglomerations of subpopulations of cells that show widely varying sensitivities toward the actions of a particular chemotherapeutic agent [104, 107]. Coupled with the fact that growing tumors also exhibit considerable evolutionary ability to enhance cell survival in an environment that is becoming hostile, this leads to multi-drug resistance of some strains of the cells. Naturally, it makes sense to combine drugs with different activation mechanisms to reach a larger population of the tumor cells—and this is what is being done—but the sad fact remains that some cells develop multi-drug resistance to a wide variety of even structurally unrelated drugs. There may even exist subpopulations of cells that are not sensitive to the treatment from the beginning (ab initio, intrinsic resistance). For certain types of cancer cells, there are simply no effective agents known.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 44.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 59.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 59.99
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

Institutional subscriptions

References

  1. K. Alitalo, Amplification of cellular oncogenes in cancer cells, Trends Biochemical Science, 10 (1985), pp. 194–197.

    Article  Google Scholar 

  2. J. Bellmunt, J.M. Trigo, E. Calvo, J. Carles, J.L. Pérez-Garcia, J.A. Virizuela, R. Lopez, M. Lázaro and J. Albanell, Activity of a multi-targeted chemo-switch regimen (sorafenib, gemcitabine, and metronomic capecitabine) in metastatic renal-cell carcinoma: a phase-2 study (SOGUG-02-06), Lancet Oncology, 2010.

    Google Scholar 

  3. A.J. Coldman and J.H. Goldie, A model for the resistance of tumor cells to cancer chemotherapeutic agents, Mathematical Biosciences, 65, (1983), pp. 291–307.

    Article  MATH  Google Scholar 

  4. A.J. Coldman and J.H. Goldie, A stochastic model for the origin and treatment of tumors containing drug-resistant cells, Bulletin of Mathematical Biology, 48, (1986), pp. 279–292.G

    Google Scholar 

  5. M.I.S. Costa, J.L. Boldrini and R.C. Bassanezi, Drug kinetics and drug resistance in optimal chemotherapy, Mathematical Biosciences, 125, (1995), pp. 191–209.

    Article  MATH  Google Scholar 

  6. M. Eisen, Mathematical Models in Cell Biology and Cancer Chemotherapy, Lecture Notes in Biomathematics, Vol. 30, Springer Verlag, Berlin, 1979.

    Book  Google Scholar 

  7. R.A. Gatenby, A.S. Silva, R.J. Gillies, and B.R. Frieden, Adaptive therapy, Cancer Research, 69, 4894–4903, (2009).

    Article  Google Scholar 

  8. H. Gardner-Moyer, Sufficient conditions for a strong minimum in singular control problems, SIAM J. Control, 11 (1973), pp. 620–636.

    Article  MathSciNet  Google Scholar 

  9. J.H. Goldie, Drug resistance in cancer: a perspective, Cancer and Metastasis Review, 20, (2001), pp. 63–68.

    Article  Google Scholar 

  10. J.H. Goldie and A. Coldman, A model for resistance of tumor cells to cancer chemotherapeutic agents, Mathematical Biosciences, 65, (1983), pp. 291–307.

    Article  MATH  Google Scholar 

  11. J.H. Goldie and A. Coldman, Quantitative model for multiple levels of drug resistance in clinical tumors, Cancer Treatment Reports, 67, (1983), pp. 923–931.

    Google Scholar 

  12. J.H. Goldie and A. Coldman, Drug Resistance in Cancer, Cambridge University Press, 1998.

    Google Scholar 

  13. R. Goodman, Introduction to Stochastic Models, Benjamin Cummings, Menlo Park, CA, 1988.

    Google Scholar 

  14. R. Grantab, S. Sivananthan and I.F. Tannock, The penetration of anticancer drugs through tumor tissue as a function of cellular adhesion and packing density of tumor cells, Cancer Research, 66, (2006), pp. 1033–1039.

    Article  Google Scholar 

  15. J. Greene, O. Lavi, M.M. Gottesman and D. Levy, The impact of cell density and mutations in a model of multidrug resistance in solid tumors, Bulletin of Mathematical Biology, 74, (2014), pp. 627–653, doi:10.1007/s11538-014-9936-8.

    Article  MathSciNet  Google Scholar 

  16. P. Hahnfeldt and L. Hlatky, Cell resensitization during protracted dosing of heterogeneous cell populations, Radiation Research, 150, (1998), pp. 681–687.

    Article  Google Scholar 

  17. L.E. Harnevo and Z. Agur, The dynamics of gene amplification described as a multitype compartmental model and as a branching process, Mathematical Biosciences, 103 (1991), pp. 115–138.

    Article  MATH  MathSciNet  Google Scholar 

  18. L.E. Harnevo and Z. Agur, Drug resistance as a dynamic process in a model for multistep gene amplification under various levels of selection stringency, Cancer Chemotherapy and Pharmacology, 30, (1992), pp. 469–476.

    Article  Google Scholar 

  19. T.L. Jackson and H. Byrne, A mathematical model to study the effects of drug resistance and vascularization on the response of solid tumors to chemotherapy, Mathematical Biosciences, 164, (2000), pp. 17–38.

    Article  MATH  MathSciNet  Google Scholar 

  20. S. Karlin and H.M. Taylor, A First Course in Stochastic Processes, Academic Press, San Diego, 1975.

    MATH  Google Scholar 

  21. M. Kimmel and D.E. Axelrod, Mathematical models of gene amplification with applications to cellular drug resistance and tumorigenicity, Genetics, 125 (1990), pp. 633–644.

    Google Scholar 

  22. M. Kimmel and A. Swierniak, Control theory approach to cancer chemotherapy: benefiting from phase dependence and overcoming drug resistance, in: Tutorials in Mathematical Biosciences III: Cell Cycle, Proliferation, and Cancer, A. Friedman, ed., Lecture Notes in Mathematics, Vol. 1872, Springer, New York, (2006), pp. 185-221.

    Google Scholar 

  23. O. Lavi, J. Greene, D. Levy, and M. Gottesman, The role of cell density and intratumoral heterogeneity in multidrug resistance, Cancer Research, 73, (2013), pp. 7168–7175.

    Article  Google Scholar 

  24. U. Ledzewicz and H. Schättler, Drug resistance in cancer chemotherapy as an optimal control problem, Discrete and Continuous Dynamical Systems, Series B, 6, (2006), pp. 129–150.

    Google Scholar 

  25. U. Ledzewicz, H. Schättler, M. Reisi Gahrooi and S. Mahmoudian Dehkordi, On the MTD paradigm and optimal control for multi-drug cancer chemotherapy, Mathematical Biosciences and Engineering (MBE), 10(3), (2013), pp. 803–819, doi:10.3934/mbe.2013.10.803.

    Google Scholar 

  26. L.A. Loeb, A mutator phenotype in cancer, Cancer Research, 61, (2001), pp. 3230–3239.

    Google Scholar 

  27. A. Lorz, T. Lorenzi, M.E. Hochberg, J. Clairambault and B. Perthame, Population adaptive evolution, chemotherapeutic resistance and multiple anti-cancer therapies, ESAIM: Mathematical Modelling and Numerical Analysis, 47, (2013) pp. 377-399, doi:10.1051/m2an/2012031.

    Article  MATH  MathSciNet  Google Scholar 

  28. A. Lorz, T. Lorenzi, J. Clairambault, A. Escargueil and B. Perthame, Effects of space structure and combination therapies on phenotypic heterogeneity and drug resistance in solid tumors, preprint

    Google Scholar 

  29. R.B. Martin, Optimal control drug scheduling of cancer chemotherapy, Automatica, 28, (1992), pp. 1113–1123.

    Article  Google Scholar 

  30. L. Norton and R. Simon, Growth curve of an experimental solid tumor following radiotherapy, J. of the National Cancer Institute, 58, (1977), pp. 1735–1741.

    Google Scholar 

  31. L. Norton and R. Simon, Tumor size, sensitivity to therapy, and design of treatment schedules, Cancer Treatment Reports, 61, (1977), pp. 1307–1317.

    Google Scholar 

  32. L. Norton and R. Simon, The Norton-Simon hypothesis revisited, Cancer Treatment Reports, 70, (1986), pp. 41–61.

    Google Scholar 

  33. K. Pietras and D. Hanahan, A multi-targeted, metronomic and maximum tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer, J. of Clinical Oncology, 23, (2005), pp. 939–952.

    Article  Google Scholar 

  34. R.T. Schimke, Gene amplification, drug resistance and cancer, Cancer Research, 44, (1984), pp. 1735–1742.

    Google Scholar 

  35. A. Swierniak, A. Polanski, M. Kimmel, A. Bobrowski and J. Smieja, Qualitative analysis of controlled drug resistance model - inverse Laplace and semigroup approach, Control and Cybernetics, 28, (1999), pp. 61–75.

    MATH  Google Scholar 

  36. A. Swierniak and J. Smieja, Cancer chemotherapy optimization under evolving drug resistance, Nonlinear Analysis, 47, (2000), pp. 375–386.

    Article  MathSciNet  Google Scholar 

  37. C. Tomasetti and D. Levy, An elementary approach to modeling drug resistance in cancer, Mathematical Biosciences and Engineering, 7, (2010), pp. 905–918.

    Article  MATH  MathSciNet  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Schättler, H., Ledzewicz, U. (2015). Cancer Chemotherapy for Heterogeneous Tumor Cell Populations and Drug Resistance. In: Optimal Control for Mathematical Models of Cancer Therapies. Interdisciplinary Applied Mathematics, vol 42. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-2972-6_3

Download citation

Publish with us

Policies and ethics