Numerical Study of Therapeutic Effectiveness of Bolus Injection and Continuous Infusion on Drug Delivery to Vascularized Solid Tumor

Document Type : Research Article

Authors

1 Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran

2 Department of Mechanical Engineering, K N Toosi University of Technology, Tehran, Iran

Abstract

Effective delivery of drugs to tumor cells is essential for the success of most anticancer therapies. In this study, two-dimensional modeling for spatiotemporal distribution of doxorubicin concentration under bolus injection and the continuous infusion is presented. Mathematical simulations have been performed considering the main physical and biochemical processes in drug delivery to tumor cells. Anticancer effectiveness is evaluated through changes in tumor cell density based on predicted intracellular concentrations. Unlike most computational models, which assume a uniform distribution of blood vessels in the tumor, the vascular network is produced using a sprouting angiogenesis method. The results demonstrate that the drugs accumulate more in areas with high vascular density, resulting in improved drug cytotoxicity. Compared to bolus injection, continuous infusion leads to longer high level maintenance of intracellular drug concentrations in the tumor, which is more effective in improving the cytotoxic effect. Although bolus injection leads to a 90% higher extracellular concentration peak, there is the risk of severe side effects. Also, continuous infusion by keeping doxorubicin at a higher level in the tumor leads to improved anticancer effectiveness by about 26% relative to the effectiveness of bolus injection at the end of the treatment.
 

Keywords


[1] W. Globocan, Estimated cancer incidence, mortality and prevalence worldwide in 2012, Int Agency Res Cancer,  (2012).
[2] M. Souri, M. Soltani, F.M. Kashkooli, M.K. Shahvandi, Engineered strategies to enhance tumor penetration of drug-loaded nanoparticles, Journal of Controlled Release, 341 (2022) 227-246.
[3] M. Souri, M. Soltani, F.M. Kashkooli, M.K. Shahvandi, M. Chiani, F.S. Shariati, M.R. Mehrabi, L.L. Munn, Towards principled design of cancer nanomedicine to accelerate clinical translation, Materials Today Bio,  (2022) 100208.
[4] M. Aghamirsalim, M. Mobaraki, M. Soltani, M. Kiani Shahvandi, M. Jabbarvand, E. Afzali, K. Raahemifar, 3D Printed Hydrogels for Ocular Wound Healing, Biomedicines, 10(7) (2022) 1562.
[5] F.M. Kashkooli, M. Soltani, M. Souri, C. Meaney, M. Kohandel, Nexus between in silico and in vivo models to enhance clinical translation of nanomedicine, Nano Today, 36 (2021) 101057.
[6] F.M. Kashkooli, M. Soltani, M. Souri, Controlled anti-cancer drug release through advanced nano-drug delivery systems: Static and dynamic targeting strategies, Journal of controlled release, 327 (2020) 316-349.
[7] J.Y. Oh, H.S. Kim, L. Palanikumar, E.M. Go, B. Jana, S.A. Park, H.Y. Kim, K. Kim, J.K. Seo, S.K. Kwak, Cloaking nanoparticles with protein corona shield for targeted drug delivery, Nature communications, 9(1) (2018) 1-9.
[8] M. Soltani, F. Moradi Kashkooli, M. Souri, S. Zare Harofte, T. Harati, A. Khadem, M. Haeri Pour, K. Raahemifar, Enhancing clinical translation of cancer using nanoinformatics, Cancers, 13(10) (2021) 2481.
[9] D. Gewirtz, A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin, Biochemical pharmacology, 57(7) (1999) 727-741.
[10] P.K. Singal, N. Iliskovic, Doxorubicin-induced cardiomyopathy, New England Journal of Medicine, 339(13) (1998) 900-905.
[11] W.M. Saltzman, Drug delivery: engineering principles for drug therapy, Oxford University Press, 2001.
[12] S.S. Legha, R.S. BENJAMIN, B. MACKAY, M. EWER, S. WALLACE, M. VALDIVIESO, S.L. RASMUSSEN, G.R. BLUMENSCHEIN, E.J. FREIREICH, Reduction of doxorubicin cardiotoxicity by prolonged continuous intravenous infusion, Annals of internal medicine, 96(2) (1982) 133-139.
[13] G. Hortobagyi, D. Frye, A. Buzdar, M. Ewer, G. Fraschini, V. Hug, F. Ames, E. Montague, C. Carrasco, B. Mackay, Decreased cardiac toxicity of doxorubicin administered by continuous intravenous infusion in combination chemotherapy for metastatic breast carcinoma, Cancer, 63(1) (1989) 37-45.
[14] A.W. El-Kareh, T.W. Secomb, A mathematical model for comparison of bolus injection, continuous infusion, and liposomal delivery of doxorubicin to tumor cells, Neoplasia, 2(4) (2000) 325-338.
[15] L.T. Baxter, R.K. Jain, Transport of fluid and macromolecules in tumors. I. Role of interstitial pressure and convection, Microvascular research, 37(1) (1989) 77-104.
[16] R.K. Jain, L.T. Baxter, Mechanisms of heterogeneous distribution of monoclonal antibodies and other macromolecules in tumors: significance of elevated interstitial pressure, Cancer research, 48(24_Part_1) (1988) 7022-7032.
[17] M. Soltani, P. Chen, Numerical modeling of fluid flow in solid tumors, PloS one, 6(6) (2011) e20344.
[18] M. Soltani, P. Chen, Effect of tumor shape and size on drug delivery to solid tumors, Journal of biological engineering, 6(1) (2012) 1-15.
[19] L. Akkari, A. Lujambio, Role of Tumor Microenvironment in Hepatocellular Carcinoma Resistance, in:  Resistance to Molecular Therapies for Hepatocellular Carcinoma, Springer, 2017, pp. 45-64.
[20] J. Folkman, M. Klagsbrun, Angiogenic factors, Science, 235(4787) (1987) 442-447.
[21] T. Anada, J. Fukuda, Y. Sai, O. Suzuki, An oxygen-permeable spheroid culture system for the prevention of central hypoxia and necrosis of spheroids, Biomaterials, 33(33) (2012) 8430-8441.
[22] L. Jakobsson, C.A. Franco, K. Bentley, R.T. Collins, B. Ponsioen, I.M. Aspalter, I. Rosewell, M. Busse, G. Thurston, A. Medvinsky, Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting, Nature cell biology, 12(10) (2010) 943-953.
[23] H. Hashizume, P. Baluk, S. Morikawa, J.W. McLean, G. Thurston, S. Roberge, R.K. Jain, D.M. McDonald, Openings between defective endothelial cells explain tumor vessel leakiness, The American journal of pathology, 156(4) (2000) 1363-1380.
[24] M.R. Junttila, F.J. De Sauvage, Influence of tumour micro-environment heterogeneity on therapeutic response, Nature, 501(7467) (2013) 346-354.
[25] P. Carmeliet, R.K. Jain, Angiogenesis in cancer and other diseases, nature, 407(6801) (2000) 249-257.
[26] R.K. Jain, R.T. Tong, L.L. Munn, Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: insights from a mathematical model, Cancer research, 67(6) (2007) 2729-2735.
[27] K.-A. Norton, A.S. Popel, Effects of endothelial cell proliferation and migration rates in a computational model of sprouting angiogenesis, Scientific reports, 6(1) (2016) 1-10.
[28] A.R. Anderson, M.A. Chaplain, S. McDougall, A hybrid discrete-continuum model of tumour induced angiogenesis, in:  Modeling Tumor Vasculature, Springer, 2012, pp. 105-133.
[29] F. Milde, M. Bergdorf, P. Koumoutsakos, A hybrid model for three-dimensional simulations of sprouting angiogenesis, Biophysical journal, 95(7) (2008) 3146-3160.
[30] M. Souri, F. Moradi Kashkooli, M. Soltani, Analysis of magneto-hyperthermia duration in nano-sized drug delivery system to solid tumors using intravascular-triggered thermosensitive-liposome, Pharmaceutical Research, 39(4) (2022) 753-765.
[31] M. Kiani Shahvandi, M. Soltani, F. Moradi Kashkooli, B. Saboury, A. Rahmim, Spatiotemporal multi-scale modeling of radiopharmaceutical distributions in vascularized solid tumors, Scientific reports, 12(1) (2022) 1-18.
[32] S. Eikenberry, A tumor cord model for doxorubicin delivery and dose optimization in solid tumors, Theoretical Biology and Medical Modelling, 6(1) (2009) 1-20.
[33] M. Soltani, M. Sefidgar, M. Casey, R. Wahl, R. Subramaniam, A. Rahmim, Comprehensive modeling of the spatiotemporal distribution of PET tracer uptake in solid tumors based on the Convection-Diffusion-Reaction equation, in:  2014 IEEE Nuclear Science Symposium and Medical Imaging Conference (NSS/MIC), IEEE, 2014, pp. 1-12.
[34] T. Stylianopoulos, L.L. Munn, R.K. Jain, Reengineering the physical microenvironment of tumors to improve drug delivery and efficacy: from mathematical modeling to bench to bedside, Trends in cancer, 4(4) (2018) 292-319.
[35] J.S. Lazo, K.L. Parker, L. Bruton, Goodman & Gilman's the pharmacological basis of therapeutics, McGraw-Hill Publishing, 2005.
[36] Y.-M.F. Goh, H.L. Kong, C.-H. Wang, Simulation of the delivery of doxorubicin to hepatoma, Pharmaceutical Research, 18(6) (2001) 761-770.
[37] Q. Dai, S. Wilhelm, D. Ding, A.M. Syed, S. Sindhwani, Y. Zhang, Y.Y. Chen, P. MacMillan, W.C. Chan, Quantifying the ligand-coated nanoparticle delivery to cancer cells in solid tumors, ACS nano, 12(8) (2018) 8423-8435.
[38] C. Liu, J. Krishnan, X.Y. Xu, A systems-based mathematical modelling framework for investigating the effect of drugs on solid tumours, Theoretical Biology and Medical Modelling, 8(1) (2011) 1-21.
[39] C. Voutouri, N.D. Kirkpatrick, E. Chung, F. Mpekris, J.W. Baish, L.L. Munn, D. Fukumura, T. Stylianopoulos, R.K. Jain, Experimental and computational analyses reveal dynamics of tumor vessel cooption and optimal treatment strategies, Proceedings of the National Academy of Sciences, 116(7) (2019) 2662-2671.
[40] Y. Boucher, R.K. Jain, Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse, Cancer research, 52(18) (1992) 5110-5114.
[41] T.W. Sheu, M.A. Solovchuk, A.W. Chen, M. Thiriet, On an acoustics–thermal–fluid coupling model for the prediction of temperature elevation in liver tumor, International Journal of Heat and Mass Transfer, 54(17-18) (2011) 4117-4126.
[42] L.T. Baxter, R.K. Jain, Transport of fluid and macromolecules in tumors. IV. A microscopic model of the perivascular distribution, Microvascular research, 41(2) (1991) 252-272.
[43] M.B. Wolf, P.D. Watson, D. Scott 2nd, Integral-mass balance method for determination of solvent drag reflection coefficient, American Journal of Physiology-Heart and Circulatory Physiology, 253(1) (1987) H194-H204.
[44] S. Eikenberry, A tumor cord model for doxorubicin delivery and dose optimization in solid tumors, Theoretical Biology and Medical Modelling, 6(1) (2009) 16.
[45] R.E. Eliaz, S. Nir, C. Marty, F.C. Szoka, Determination and modeling of kinetics of cancer cell killing by doxorubicin and doxorubicin encapsulated in targeted liposomes, Cancer research, 64(2) (2004) 711-718.
[46] C. Liu, J. Krishnan, X.Y. Xu, Investigating the effects of ABC transporter-based acquired drug resistance mechanisms at the cellular and tissue scale, Integrative Biology, 5(3) (2013) 555-568.
[47] M. Soltani, M. Souri, F. Moradi Kashkooli, Effects of hypoxia and nanocarrier size on pH-responsive nano-delivery system to solid tumors, Scientific Reports, 11(1) (2021) 1-12.
[48] M. Souri, M. Soltani, F. Moradi Kashkooli, Computational modeling of thermal combination therapies by magneto-ultrasonic heating to enhance drug delivery to solid tumors, Scientific reports, 11(1) (2021) 1-12.
[49] W. Zhan, X.Y. Xu, A mathematical model for thermosensitive liposomal delivery of doxorubicin to solid tumour, Journal of drug delivery, (2013) 172529.
[50] T. Stylianopoulos, R.K. Jain, Combining two strategies to improve perfusion and drug delivery in solid tumors, Proceedings of the National Academy of Sciences, 110(46) (2013) 18632-18637.
[51] M. Soltani, P. Chen, Numerical modeling of interstitial fluid flow coupled with blood flow through a remodeled solid tumor microvascular network, PloS one, 8(6) (2013) e67025.
[52] T. Hompland, C. Ellingsen, K.M. Øvrebø, E.K. Rofstad, Interstitial fluid pressure and associated lymph node metastasis revealed in tumors by dynamic contrast-enhanced MRI, Cancer research, 72(19) (2012) 4899-4908.