Skip to main content

Advertisement

Log in

Mechanistic Pharmacokinetic/Pharmacodynamic Model of Sunitinib and Dopamine in MCF-7/Adr Xenografts: Linking Cellular Heterogeneity to Tumour Burden

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

The self-renewal and differentiation of cancer stem-like cells (CSCs) leads to cellular heterogeneity, causing one of the greatest challenges in cancer therapy. Growing evidence suggests that CSC-targeting therapy enhances the effect of concomitant antitumour therapy. To gain an in-depth understanding of this enhanced effect, the kinetic profile of estimated CSC frequency (the fraction of CSCs in tumour) was evaluated for in vivo characterization of cellular heterogeneity using sunitinib and dopamine as a paradigm combination therapy. Female MCF-7/Adr xenografted Balb/c nude mice were treated with sunitinib (p.o., 20 mg/kg) and dopamine (i.p., 50 mg/kg), alone or in combination. Estimated CSC frequency and tumour size were measured over time. Mechanistic PK/PD modelling was performed to quantitatively describe the relationship between drug concentration, estimated CSC frequency and tumour size. Sunitinib reduced tumour size by inducing apoptosis of differentiated tumour cells (DTCs) and enriched CSCs by stimulating its proliferation. Dopamine exhibited anti-CSC effects by suppressing the capacity of CSCs and inducing its differentiation. Simulation and animal studies indicated that concurrent administration was superior to sequential administration under current experimental conditions. Alongside tumour size, the current study provides mechanistic insights into the estimation of CSC frequency as an indicator for cellular heterogeneity. This forms the conceptual basis for in vivo characterization of other combination therapies in preclinical cancer studies.

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

Similar content being viewed by others

References

  1. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501(7467):328–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. Cancer stem cells--perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res. 2006;66(19):9339–44.

    Article  CAS  PubMed  Google Scholar 

  3. Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol. 2005;45(8):872–7.

    Article  CAS  PubMed  Google Scholar 

  4. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008;100(9):672–9.

    Article  CAS  PubMed  Google Scholar 

  5. Brooks MD, Burness ML, Wicha MS. Therapeutic implications of cellular heterogeneity and plasticity in breast cancer. Cell Stem Cell. 2015;17(3):260–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ablett MP, Singh JK, Clarke RB. Stem cells in breast tumours: are they ready for the clinic? Eur J Cancer. 2012;48(14):2104–16.

    Article  CAS  PubMed  Google Scholar 

  7. Wang S, Mou Z, Ma Y, Li J, Li J, Ji X, et al. Dopamine enhances the response of sunitinib in the treatment of drug-resistant breast cancer: involvement of eradicating cancer stem-like cells. Biochem Pharmacol. 2015;95(2):98–109.

    Article  CAS  PubMed  Google Scholar 

  8. Hao F, Wang S, Zhu X, Xue J, Li J, Wang L, et al. Pharmacokinetic-pharmacodynamic modeling of the anti-tumor effect of sunitinib combined with dopamine in the human non-small cell lung Cancer Xenograft. Pharm Res. 2017;34(2):408–18.

    Article  CAS  PubMed  Google Scholar 

  9. Christensen JG. A preclinical review of sunitinib, a multitargeted receptor tyrosine kinase inhibitor with anti-angiogenic and antitumour activities. Ann Oncol. 2007;18 Suppl 10(suppl_10):x3–10.

    Article  CAS  PubMed  Google Scholar 

  10. Roskoski R Jr. Sunitinib: a VEGF and PDGF receptor protein kinase and angiogenesis inhibitor. Biochem Biophys Res Commun. 2007;356(2):323–8.

    Article  CAS  PubMed  Google Scholar 

  11. Conley SJ, Gheordunescu E, Kakarala P, Newman B, Korkaya H, Heath AN, et al. Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia. Proc Natl Acad Sci U S A. 2012;109(8):2784–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chinchar E, Makey K, Gibson J, Chen F, Cole S, Megason G, et al. Sunitinib significantly suppresses the proliferation, migration, apoptosis resistance, tumor angiogenesis and growth of triple-negative breast cancers but increases breast cancer stem cells. Vasc Cell. 2014;6(1):1–12.

    Article  Google Scholar 

  13. Mackey JR, Kerbel RS, Gelmon KA, McLeod DM, Chia SK, Rayson D, et al. Controlling angiogenesis in breast cancer: a systematic review of anti-angiogenic trials. Cancer Treat Rev. 2012;38(6):673–88.

    Article  CAS  PubMed  Google Scholar 

  14. Sachlos E, Risueno RM, Laronde S, Shapovalova Z, Lee JH, Russell J, et al. Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell. 2012;149(6):1284–97.

    Article  CAS  PubMed  Google Scholar 

  15. Ribba B, Holford NH, Magni P, Trocóniz I, Gueorguieva I, Girard P, et al. A review of mixed-effects models of tumor growth and effects of anticancer drug treatment used in population analysis. CPT Pharmacometrics Syst Pharmacol. 2014;3(5):1–10.

    Article  Google Scholar 

  16. Ricardo S, Vieira AF, Gerhard R, Leitao D, Pinto R, Cameselle-Teijeiro JF, et al. Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol. 2011;64(11):937–46.

    Article  PubMed  Google Scholar 

  17. Beal S, Sheiner L, Boeckmann A, Bauer R. NONMEM User’s Guides. Ellicott City: Icon Development Solutions; 2009. p. 2009.

    Google Scholar 

  18. Keizer RJ, Karlsson MO, Hooker A. Modeling and simulation workbench for NONMEM: tutorial on Pirana, PsN, and Xpose. CPT Pharmacometrics Syst Pharmacol. 2013;2(6):e50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yan Y, Jiang W, Liu L, Wang X, Ding C, Tian Z, et al. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell. 2015;160(1–2):62–73.

    Article  CAS  PubMed  Google Scholar 

  20. Li J, Li J, Wang S, Yuan Y, Su Q, Lu W, et al. Simultaneous determination of sunitinib and its active metabolites N-desethyl sunitinib (SU12662) in nude mice plasma by liquid chromatography tandem mass spectrometry and its application to a pharmacokinetic study. J Chin Pharm Sci. 2015;24(4):217–24.

    CAS  Google Scholar 

  21. Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol. 2006;24(1):25–35.

    Article  CAS  PubMed  Google Scholar 

  22. Martelotto LG, Ng CK, Piscuoglio S, Weigelt B, Reis-Filho JS. Breast cancer intra-tumor heterogeneity. Breast Cancer Res. 2014;16(3):210.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ganguly R, Puri IK. Mathematical model for the cancer stem cell hypothesis. Cell Prolif. 2006;39(1):3–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Koch G, Walz A, Lahu G, Schropp J. Modeling of tumor growth and anticancer effects of combination therapy. J Pharmacokinet Pharmacodyn. 2009;36(2):179–97.

    Article  CAS  PubMed  Google Scholar 

  25. Simeoni M, Magni P, Cammia C, De Nicolao G, Croci V, Pesenti E, et al. Predictive pharmacokinetic-pharmacodynamic modeling of tumor growth kinetics in xenograft models after administration of anticancer agents. Cancer Res. 2004;64(3):1094–101.

    Article  CAS  PubMed  Google Scholar 

  26. Al Faraj A, Shaik AS, Al Sayed B, Halwani R, Al JI. Specific targeting and noninvasive imaging of breast cancer stem cells using single-walled carbon nanotubes as novel multimodality nanoprobes. Nanomed (London, England). 2016;11(1):31–46.

    Article  Google Scholar 

  27. Ganguly R, Puri IK. Mathematical model for chemotherapeutic drug efficacy in arresting tumour growth based on the cancer stem cell hypothesis. Cell Prolif. 2007;40(3):338–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Molina-Pena R, Alvarez MM. A simple mathematical model based on the cancer stem cell hypothesis suggests kinetic commonalities in solid tumor growth. PLoS One. 2012;7(2):e26233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ma YH, Wang SY, Ren YP, Li J, Guo TJ, Lu W, et al. Antitumor effect of axitinib combined with dopamine and PK-PD modeling in the treatment of human breast cancer xenograft. Acta Pharmacol Sin. 2018;40(2):243–56.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Karlsson MO, Jonsson EN, Wiltse CG, Wade JR. Assumption testing in population pharmacokinetic models: illustrated with an analysis of moxonidine data from congestive heart failure patients. J Pharmacokinet Biopharm. 1998;26(2):207–46.

    Article  CAS  PubMed  Google Scholar 

  31. Workgroup EM, Marshall SF, Burghaus R, Cosson V, Cheung SY, Chenel M, et al. Good practices in model-informed drug discovery and development: practice, application, and documentation. CPT Pharmacometrics Syst Pharmacol. 2016;5(3):93–122.

    Article  Google Scholar 

  32. Ooi Q-X, Wright DF, Isbister GK, Duffull SB. Evaluation of assumptions underpinning pharmacometric models. AAPS J. 2019;21(5):97.

    Article  PubMed  Google Scholar 

  33. Noori S, Friedlich P, Seri I. Pharmacology review developmentally regulated cardiovascular, renal, and neuroendocrine effects of dopamine. NeoReviews. 2003;4(10):e283–e8.

    Article  Google Scholar 

  34. Felip E, Massuti B, Camps C, Benito D, Isla D, Gonzalez-Larriba JL, et al. Superiority of sequential versus concurrent administration of paclitaxel with etoposide in advanced non-small cell lung cancer: comparison of two phase II trials. Clin Cancer Res. 1998;4(11):2723–8.

    CAS  PubMed  Google Scholar 

  35. Perez EA, Suman VJ, Davidson NE, Gralow JR, Kaufman PA, Visscher DW, et al. Sequential versus concurrent trastuzumab in adjuvant chemotherapy for breast cancer. J Clin Oncol. 2011;29(34):4491–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5(4):275–84.

    Article  CAS  PubMed  Google Scholar 

  37. Sarkar C, Chakroborty D, Chowdhury UR, Dasgupta PS, Basu S. Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin Cancer Res. 2008;14(8):2502–10.

    Article  CAS  PubMed  Google Scholar 

  38. Zhang L, Yao HJ, Yu Y, Zhang Y, Li RJ, Ju RJ, et al. Mitochondrial targeting liposomes incorporating daunorubicin and quinacrine for treatment of relapsed breast cancer arising from cancer stem cells. Biomaterials. 2012;33(2):565–82.

    Article  CAS  PubMed  Google Scholar 

  39. Liu S, Wicha MS. Targeting breast cancer stem cells. J Clin Oncol. 2010;28(25):4006–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14(3):275–91.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Prof. Stephen Duffull (Otago Pharmacometrics Group, University of Otago) for his careful reading of the manuscript.

Funding

This study is sponsored by National Natural Science Foundation of China (NSFC) [Grant 81473277 and 81703605].

Author information

Authors and Affiliations

Authors

Contributions

S.W. performed experiments and wrote the paper. X.Z analysed the data and wrote the paper. M. H. and F.H. performed experiments. W.L. designed experiments. T.Z. designed experiments and wrote the paper.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

ESM 1

(DOCX 505 kb)

Appendices

Appendices

Existing Knowledge

Here, we summarise the existing knowledge of CSC theory and the related pharmacological activities of dopamine and sunitinib. All information was incorporated into a mechanistic model that described the sunitinib/dopamine combination in the treatment of drug-resistant breast cancer:

  1. 1)

    The cancer stem-like cells (CSCs) and the differentiated tumour cells (DTCs) own the capacity of converting to the other one [1].

  2. 2)

    The size of CSCs is much smaller than DTCs [38].

  3. 3)

    The DTC population grow more aggressively and mainly contribute to the tumour bulk [39].

  4. 4)

    The CSC population cycle very slowly and eventually reach the steady state during the evolution of tumour [23, 40].

  5. 5)

    Dopamine can eradicate CSCs [7].

  6. 6)

    Sunitinib is a multi-targeted receptor tyrosine kinase inhibitor (TKI) with anti-angiogenic and antitumour activities [9, 10].

  7. 7)

    Sunitinib enriches the CSC population within the tumour [11,12,13].

Derivation of the Equation for Tumour Volume

In this study, the tumour volume is approximated by the sum of volumes of different cancel cells. As the tumour mainly consists of two representative cell subpopulations (i.e. CSCs and DTCs), the tumour volumes at time 0 (Eq. A1) and t (Eq. A2) are derived:

$$ {V}_0={CSC}_0\cdotp {V}_{s- CSC}+{DTC}_0\cdotp {V}_{s- DTC} $$
(A1)
$$ V(t)= CSC(t)\cdotp {V}_{s- CSC}+ DTC(t)\cdotp {V}_{s- DTC} $$
(A2)

Here, V0 and V(t) represent the tumour volume at time 0 and t. CSC(t) and DTC(t) denote the amount of CSCs or DTCs at time t, respectively. Vs − CSC and Vs − DTC represent the volume of a single cell of CSC or DTC, respectively.

Re-organizing the Eqs. A1 and A2 yields the expression of tumour volume at time t related to the tumour volume at time 0 (Eq. A3):

$$ V(t)={V}_0\cdotp \frac{CSC(t)\cdotp {V}_{s- CSC}+ DTC(t)\cdotp {V}_{s- DTC}}{CSC_0\cdotp {V}_{s- CSC}+{DTC}_0\cdotp {V}_{s- DTC}} $$
(A3)

Dividing the numerator and denominator by Vs − DTC yields Eq. A4:

$$ V(t)={V}_0\cdotp \frac{CSC(t)\cdotp Ratio+ DTC(t)}{CSC_0\cdotp Ratio+{DTC}_0} $$
(A4)

Here, Ratio represents the volume ratio of a single CSC to a single DTC.

Since the exact amount of initial tumour cells is unknown, the initial amount of total tumour cells is assumed to be 100 (arbitrary units). From our experiments, the average estimated CSC frequency before the first drug administration is 1.55%. Thus, the initial amount of CSCs (CSC0) is 1.55 units, and the initial amount of DTCs (DTC0) is 98.45 units. In addition, it is known that the size of CSCs is much smaller than DTCs (Existing knowledge 2), indicating that the volume ratio of a single CSC to a single DTC (Ratio) is much smaller than 1. Therefore, the product of CSC0 and Ratio (CSC0 ∙ Ratio) is much smaller than DTC0. Thus, Eq. A4 can be reduced into Eq. A5.

$$ V(t)={V}_0\cdotp \frac{CSC(t)\cdotp Ratio+ DTC(t)}{DTC_0} $$
(A5)

It is known that the DTC population grow more aggressively and mainly contribute to the tumour bulk (Existing knowledge 3), indicating that DTC(t) is much larger than CSC(t). Furthermore, Ratio is much smaller than 1 (Existing knowledge 2). Hence, the product of CSC(t) and Ratio is much smaller than DTC(t). Thus, Eq. A5 can be reduced into the final expression of tumour volume (Eq. A6):

$$ V(t)={V}_0\cdotp \frac{DTC(t)}{DTC_0} $$
(A6)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, S., Zhu, X., Han, M. et al. Mechanistic Pharmacokinetic/Pharmacodynamic Model of Sunitinib and Dopamine in MCF-7/Adr Xenografts: Linking Cellular Heterogeneity to Tumour Burden. AAPS J 22, 45 (2020). https://doi.org/10.1208/s12248-020-0428-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-020-0428-5

Key Words

Navigation