Skip to main content
Log in

Modeling the Dynamics of Heterogeneity of Solid Tumors in Response to Chemotherapy

  • Original Article
  • Published:
Bulletin of Mathematical Biology Aims and scope Submit manuscript

Abstract

In this paper, we extend the model of the dynamics of drug resistance in a solid tumor that was introduced by Lorz et al. (Bull Math Biol 77:1–22, 2015). Similarly to the original, radially symmetric model, the quantities we follow depend on a phenotype variable that corresponds to the level of drug resistance. The original model is modified in three ways: (i) We consider a more general growth term that takes into account the sensitivity of resistance level to high drug dosage. (ii) We add a diffusion term in space for the cancer cells and adjust all diffusion terms (for the nutrients and for the drugs) so that the permeability of the resource and drug is limited by the cell concentration. (iii) We add a mutation term with a mutation kernel that corresponds to mutations that occur regularly or rarely. We study the dynamics of the emerging resistance of the cancer cells under continuous infusion and on–off infusion of cytotoxic and cytostatic drugs. While the original Lorz model has an asymptotic profile in which the cancer cells are either fully resistant or fully sensitive, our model allows the emergence of partial resistance levels. We show that increased drug concentrations are correlated with delayed relapse. However, when the cancer relapses, more resistant traits are selected. We further show that an on–off drug infusion also selects for more resistant traits when compared with a continuous drug infusion of identical total drug concentrations. Under certain conditions, our model predicts the emergence of a heterogeneous tumor in which cancer cells of different resistance levels coexist in different areas in space.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13
Fig. 14
Fig. 15
Fig. 16
Fig. 17
Fig. 18
Fig. 19
Fig. 20

Similar content being viewed by others

Notes

  1. Since the perturbations in the decay coefficients are small, for instance, \(\gamma _s \gg \int _0^1 p(\theta ) n(t,r,\theta ) \mathrm{d}\theta \), this estimation is similar to our simulation.

  2. The solutions can be explicitly written in terms of a modified Bessel function of the first kind \(I_0(r)\) as \(s(r) = S_1 I_0\left( \sqrt{\lambda _s}r \right) / I_0\left( \sqrt{\lambda _s} \right) \), where \(\lambda _s = {\gamma _s}/{\alpha _s}\), and similarly for \(c_1(r)\) and \(c_2(r)\). Then, \(\hat{R}_k(\theta ) = \frac{p(\theta )}{1+\mu _2 c_2(r_k)} s(r_k)\) and \( \hat{C}_k(\theta ) = \mu _1(\theta ) c_1(r_k)\).

  3. \(R_k^M = \max _{\theta } \left[ \frac{p(\theta )}{1+\mu _2 c_2(r_k)} s(r_k) \right] \), \(C_{k}^m = \min _\theta \left[ \mu _1(\theta ) c_1(r_k)\right] \). Space independent bounds can be given as \(R^M = \max _{\theta } \left[ \frac{p(\theta )}{1+\mu _2 C_2/I_0(\sqrt{\lambda _{c_2}})} S_1 \right] \) and \(C^m = \min _\theta \left[ \mu _1(\theta ) C_1/I_0(\sqrt{\lambda _{c_1}}) \right] \), where \(I_0(r)\) is a modified Bessel function of the first kind, \(\lambda _{c_1} = {\gamma _{c_1}}/{\alpha _{c_1}}\), and \(\lambda _{c_2} = {\gamma _{c_2}}/{\alpha _{c_2}}\).

  4. \(D_{KL}(Q_0||Q_t)\) is the KL divergence from the initial distribution \(Q_0(\theta ) := Q(t=0,\theta )\),

    $$\begin{aligned} D_{KL} ( Q_0 || Q_t ) := \int _0^1 Q_0(\theta ) \log \dfrac{Q_0(\theta )}{Q({t,\theta })} \mathrm{d}\theta , \end{aligned}$$

    that represents the divergence of the phenotype distribution from initial time.

  5. Similar behavior can be modeled using the Gaussian kernel (18) by considering a piecewise continuous correlation length on \(\{\varOmega _i\}\). For instance, \( \ell _u(\theta ,\theta ^{\prime }) = \ell _i( \bar{\theta } )\) on each \(\bar{\theta } \in \varOmega _i,\) where \(\ell _i( \bar{\theta } )\) is a quadratic function that decays as \(\bar{\theta }\) approaches the partition boundary.

References

  • Amir ED, Davis KL, Tadmor MD, Simonds EF, Jacob Levlne H, Bendall SC, Shenfeld DK, Krishnaswamy S, Nolan GP, Pe’er D (2013) viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol 31(6):545–552

    Article  Google Scholar 

  • Anderson AR, Chaplain M (1998) Continuous and discrete mathematical models of tumor-induced angiogenesis. Bull Math Biol 60(5):857–899

    Article  MATH  Google Scholar 

  • Anderson AR, Weaver AM, Cummings PT, Quaranta V (2006) Tumor morphology and phenotypic evolution driven by selective pressure from the microenvironment. Cell 127(5):905–915

    Article  Google Scholar 

  • Ariffin AB, Forde PF, Jahangeer S, Soden DM, Hinchion J (2014) Releasing pressure in tumors: What do we know so far and where do we go from here? Cancer Res 74:2655–62

    Article  Google Scholar 

  • Birkhead BG, Rakin EM, Gallivan S, Dones L, Rubens RD (1987) A mathematical model of the development of drug resistance to cancer chemotherapy. Eur J Cancer Clin Oncol 23:1421–1427

    Article  Google Scholar 

  • Brodie EDIII (1992) Correlational selection for color pattern and antipredator behavior in the garter snake Thamnophis ordinoides. Evolution 46:1284–1298

    Article  Google Scholar 

  • Corbett T, Griswold D, Roberts B, Peckham J, Schabel F (1975) Tumor induction relationships in development of transplantable cancers of the colon in mice for chemotherapy assays, with a note on carcinogen structure. Cancer Res 35:2434–2439

    Google Scholar 

  • de Bruin EC, Taylor TB, Swanton C (2013) Intra-tumor heterogeneity: lessons from microbial evolution and clinical implications. Genome Med 5:1–11

    Article  Google Scholar 

  • de Pillis LG, Radunskaya AE, Wiseman CL (2005) A validated mathematical model of cell-mediated immune response to tumor growth. Cancer Res 65:7950–7958

    Article  Google Scholar 

  • de Pillis LG, Savage H, Radunskaya AE (2014) Mathematical model of colorectal cancer with monoclonal antibody treatments. Br J Med Med Res 4:3101–3131

    Article  Google Scholar 

  • Fodal V, Pierobon M, Liotta L, Petricoin E (2011) Mechanisms of cell adaptation: When and how do cancer cells develop chemoresistance? Cancer J 17(2):89–95

    Article  Google Scholar 

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

    Article  Google Scholar 

  • Garvey CM, Spiller E, Lindsay D, Ct Chiang, Choi NC, Agus DB, Mallick P, Foo J, Mumenthaler SM (2016) A high-content image-based method for quantitatively studying context-dependent cell population dynamics. Sci Rep 6(29752):1–12

    Google Scholar 

  • Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P (2012) Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892

    Article  Google Scholar 

  • Gillet JP, Gottesman MM (2010) Mechanisms of multidrug resistance in cancer. Methods Mol Biol 596:47–76

    Article  Google Scholar 

  • Goldie JH, Coldman AJ (1979) A mathematical model for relating the drug sensitivity of tumors to their spontaneous mutation rate. Cancer Treat Rep 63:1727–1733

    Google Scholar 

  • Goldie JH, Coldman AJ (1983a) A model for resistance of tumor cells to cancer chemotherapeutic agents. Math Biosci 65:291–307

    Article  MATH  Google Scholar 

  • Goldie JH, Coldman AJ (1983b) Quantative model for multiple levels of drug resistance in clinical tumors. Cancer Treat Rep 67:923–931

    Google Scholar 

  • Gottesman MM (2002) Mechanisms of cancer drug resistance. Annu Rev Med 53:615–627

    Article  Google Scholar 

  • Gottesman MM, Fojo T, Bates SE (2002) Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2(1):48–58

    Article  Google Scholar 

  • Greene J, Lavi O, Gottesman MM, Levy D (2014) The impact of cell density and mutations in a model of multidrug resistance in solid tumors. Bull Math Biol 74:627–653

    Article  MathSciNet  MATH  Google Scholar 

  • Grothey A (2006) Defining the role of panitumumab in colorectal cancer. Community Oncol 3:6–10

    Google Scholar 

  • Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674

    Article  Google Scholar 

  • Iwasa Y, Nowak MA, Michor F (2006) Evolution of resistance during clonal expansion. Genetics 172:2557–2566

    Article  Google Scholar 

  • Jabin PE, Schram RS (2016) Selection-mutation dynamics with spatial dependence pp 1–21

  • Kimmel M, Swierniak A, Polanski A (1998) Infinite-dimensional model of evolution of drug resistance of cancer cells. J Math Syst Estim Control 8:1–16

    MathSciNet  MATH  Google Scholar 

  • Komarova N (2006) Stochastic modeling of drug resistance in cancer. Theor Popul Biol 239(3):351–366

    Article  MathSciNet  Google Scholar 

  • Lavi O, Gottesman MM, Levy D (2012) The dynamics of drug resistance: a mathematical perspective. Drug Resist Update 15(1–2):90–97

    Article  Google Scholar 

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

    Article  Google Scholar 

  • Lavi O, Greene J, Levy D, Gottesman MM (2014) Simplifying the complexity of resistance heterogeneity in metastatic cancer. Trend Mol Med 20:129–136

    Article  Google Scholar 

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

    Article  MathSciNet  MATH  Google Scholar 

  • Lorz A, Lorenzi T, Clairambault J, Escargueil A, Perthame B (2015) Modeling the effects of space structure and combination therapies on phenotypic heterogeneity and drug resistance in solid tumors. Bull Math Biol 77:1–22

    Article  MathSciNet  MATH  Google Scholar 

  • Medema JP (2013) Cancer stem cells: the challenges ahead. Nat Cell Biol 15(4):338–344

    Article  Google Scholar 

  • Michor F, Nowak MA, Iwasa Y (2006) Evolution of resistance to cancer therapy. Curr Pharm Des 12:261–271

    Article  Google Scholar 

  • Minchinton AI, Tannock IF (2006) Drug penetration in solid tumours. Nat Rev Cancer 6:583–592

    Article  Google Scholar 

  • Mirrahimi S, Perthame B (2015) Asymptotic analysis of a selection model with space. J Math Pures Appl 104:1108–1118

    Article  MathSciNet  MATH  Google Scholar 

  • Mumenthaler SM, Foo J, Choi NC, Heise N, Leder K, Agus DB, Pao W, Michor F, Mallick P (2015) The impact of microenvironmental heterogeneity on the evolution of drug resistance in cancer cells. Cancer Inform 14:19–31

    Google Scholar 

  • Panagiotopoulou V, Richardson G, Jensen OE, Rauch C (2010) On a biophysical and mathematical model of Pgp-mediated multidrug resistance: understanding the space-time dimension of MDR. Eur Biophys J 39:201–211

    Article  Google Scholar 

  • Panetta JC (1998) A mathematical model of drug resistance: heterogeneous tumors. Math Biosci 147:41–61

    Article  MATH  Google Scholar 

  • Rainey PB, Travisano M (1998) Adaptive radiation in a heterogeneous environment. Nature 2:69–72

    Article  Google Scholar 

  • Roose T, Chapman SJ, Maini PK (2007) Mathematical models of avascular tumor growth. SIAM Rev 49(2):179–208

    Article  MathSciNet  MATH  Google Scholar 

  • Schättler H, Ledzewicz U (2015) Optimal control for mathematical models of cancer therapies, 1st edn. Springer, Berlin

    Book  MATH  Google Scholar 

  • Simon R, Norton L (2006) The norton-simon hypothesis: designing more effective and less toxic chemotherapeutic regimens. Nat Clin Pract Oncol 3:406–407

    Article  Google Scholar 

  • Swierniak A, Kimmel M, Smieja J (2009) Mathematical modeling as a tool for planning anticancer therapy. Eur J Pharmacol 625(1–3):108–121

    Article  Google Scholar 

  • Teicher BA (2006) Cancer drug resistance. Humana Press, Totowa

    Book  Google Scholar 

  • Tomasetti C, Levy D (2010) An elementary approach to modeling drug resistance in cancer. Math Biosci Eng 7:905–918

    Article  MathSciNet  MATH  Google Scholar 

  • Trédan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 99:1441–1454

    Article  Google Scholar 

  • Vaupel P, Kallinowski F, Okunieff P (1989) Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Res 49:6449–6465

    Google Scholar 

  • Wosikowski K, Silverman JA, Bishop P, Mendelsohn J, Bates SE (2000) Reduced growth rate accompanied by aberrant epidermal growth factor signaling in drug resistant human breast cancer cells. Biochimica et Biophysica Acta 1497(2):215–226

    Article  Google Scholar 

  • Wu A, Loutherback K, Lambert G, Estévez-Salmerón L, Tlsty TD, Austin RH, Sturm JC (2013) Cell motility and drug gradients in the emergence of resistance to chemotherapy. Proc Natl Acad Sci 110(40):16,103–16,108

    Article  Google Scholar 

  • Yu P, Mustata M, Peng L, Turek JJ, Melloch MR, French PM, Nolte DD (2004) Holographic optical coherence imaging of rat osteogenic sarcoma tumor spheroids. Appl Opt 43:4862–4873

    Article  Google Scholar 

Download references

Acknowledgements

The work of DL was supported in part by the National Science Foundation under Grant Number DMS-1713109, by the John Simon Guggenheim Memorial Foundation, by the Simons Foundation, and by the Jayne Koskinas and Ted Giovanis Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Doron Levy.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cho, H., Levy, D. Modeling the Dynamics of Heterogeneity of Solid Tumors in Response to Chemotherapy. Bull Math Biol 79, 2986–3012 (2017). https://doi.org/10.1007/s11538-017-0359-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11538-017-0359-1

Keywords

Navigation