1,601
Views
10
CrossRef citations to date
0
Altmetric
Research Article

A spatiotemporal computational model of focused ultrasound heat-induced nano-sized drug delivery system in solid tumors

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Article: 2219871 | Received 13 Feb 2023, Accepted 02 May 2023, Published online: 14 Jun 2023

References

  • Ahmadi A, Hosseini-Nami S, Abed Z, et al. (2020). Recent advances in ultrasound-triggered drug delivery through lipid-based nanomaterials. Drug Discov Today 25:1–21.
  • Baxter LT, Jain RK. (1989). Transport of fluid and macromolecules in tumors. I. Role of interstitial pressure and convection. Microvasc Res 37:77–104.
  • Baxter LT, Jain RK. (1990). Transport of fluid and macromolecules in tumors. II. Role of heterogeneous perfusion and lymphatics. Microvasc Res 40:246–63.
  • Baxter LT, Jain RK. (1991). Transport of fluid and macromolecules in tumors: III. Role of binding and metabolism. Microvasc Res 41:5–23.
  • Boissenot T, Bordat A, Fattal E, Tsapis N. (2016). Ultrasound-triggered drug delivery for cancer treatment using drug delivery systems: from theoretical considerations to practical applications. J Control Release 241:144–63.
  • Boucher Y, Baxter LT, Jain RK. (1990). Interstitial pressure gradients in tissue-isolated and subcutaneous tumors: implications for therapy. Cancer Res 50:4478–84.
  • Boucher Y, Jain RK. (1992). Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse. Cancer Res 52:5110–4.
  • Butler TP, Grantham FH, Gullino PM. (1975). Bulk transfer of fluid in the interstitial compartment of mammary tumors. Cancer Res 35:3084–8.
  • Butt F. (2011). High performance computing for linear acoustic wave simulation, Ryerson University, 1–127.
  • Butt F, Abhari A, Tavakkoli J. (2011). An application of high performance computing to improve linear acoustic simulation, Conference, Spring Simulation Multi-conference, SpringSim, Boston, MA. Volume 3: Proceedings of the 14th Communications and Networking Symposium (CNS), 71–8.
  • Centelles MN, Wright M, So P-W, et al. (2018). Image-guided thermosensitive liposomes for focused ultrasound drug delivery: using NIRF-labelled lipids and topotecan to visualise the effects of hyperthermia in tumours. J Control Release 280:87–98.
  • Chabner BA, Longo DL. (2011). Cancer chemotherapy and biotherapy: principles and practice. Wolters Kluwer; Lippincott Williams & Wilkins.
  • Couture O, Foley J, Kassell NF, et al. (2014). Review of ultrasound mediated drug delivery for cancer treatment: updates from pre-clinical studies. Transl Cancer Res 3:494–511.
  • Danhier F. (2016). To exploit the tumor microenvironment: since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release 244:108–21.
  • Dewhirst MW, Secomb TW. (2017). Transport of drugs from blood vessels to tumour tissue. Nat Rev Cancer 17:738–50.
  • Dimcevski G, Kotopoulis S, Bjånes T, et al. (2016). A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release 243:172–81.
  • Du J-Z, Du X-J, Mao C-Q, Wang J. (2011). Tailor-made dual pH-sensitive polymer–doxorubicin nanoparticles for efficient anticancer drug delivery. J Am Chem Soc 133:17560–3.
  • Eikenberry S. (2009). A tumor cord model for doxorubicin delivery and dose optimization in solid tumors. Theor Biol Med Model 6:1–20.
  • Eliaz RE, Nir S, Marty C, Szoka FC. (2004). Determination and modeling of kinetics of cancer cell killing by doxorubicin and doxorubicin encapsulated in targeted liposomes. Cancer Res 64:711–8.
  • El-Kareh AW, Secomb TW. (2000). A mathematical model for comparison of bolus injection, continuous infusion, and liposomal delivery of doxorubicin to tumor cells. Neoplasia 2:325–38.
  • El-Kareh AW, Secomb TW. (2003). A mathematical model for cisplatin cellular pharmacodynamics. Neoplasia 5:161–9.
  • Filonenko E, Khokhlova V. (2001). Effect of acoustic nonlinearity on heating of biological tissue by high-intensity focused ultrasound. Acoust Phys 47:468–75.
  • Gao W, Chan JM, Farokhzad OC. (2010). pH-responsive nanoparticles for drug delivery. Mol Pharmaceut 7:1913–20.
  • Gasselhuber A, Dreher MR, Partanen A, et al. (2012). Targeted drug delivery by high intensity focused ultrasound mediated hyperthermia combined with temperature-sensitive liposomes: computational modelling and preliminary in vivo validation. Int J Hyperthermia 28:337–48.
  • Gasselhuber A, Dreher MR, Rattay F, et al. (2012). Comparison of conventional chemotherapy, stealth liposomes and temperature-sensitive liposomes in a mathematical model. Plos ONE 7:e47453.
  • Gasselhuber A, Dreher MR, Rattay F, et al. (2012). Comparison of conventional chemotherapy, stealth liposomes and temperature-sensitive liposomes in a mathematical model.
  • Goh Y-MF, Kong HL, Wang C-H. (2001). Simulation of the delivery of doxorubicin to hepatoma. Pharm Res 18:761–70.
  • Greene RF, Collins JM, Jenkins JF, et al. (1983). Plasma pharmacokinetics of adriamycin and adriamycinol: implications for the design of in vitro experiments and treatment protocols. Cancer Res 43:3417–21.
  • Grüll H, Langereis S. (2012). Hyperthermia-triggered drug delivery from temperature-sensitive liposomes using MRI-guided high intensity focused ultrasound. J Control Release 161:317–27.
  • Haemmerich D, Ramajayam KK, Newton DA. (2023). Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers 15:398.
  • Hallaj IM, Cleveland RO. (1999). FDTD simulation of finite-amplitude pressure and temperature fields for biomedical ultrasound. J Acoust Soc Am 105:L7–12.
  • Hijnen N, Kneepkens E, de Smet M, et al. (2017). Thermal combination therapies for local drug delivery by magnetic resonance-guided high-intensity focused ultrasound. Proc Natl Acad Sci U S A 114:E4802–E4811.
  • Hornsby TK, Jakhmola A, Kolios MC, Tavakkoli J. (2023). A Quantitative study of thermal and non-thermal mechanisms in ultrasound-induced nano-drug delivery. Ultrasound Med Biol 49:1288–98.
  • Hornsby T, Kashkooli FM, Jakhmola A, et al. (2022). Measuring drug release induced by thermal and non-thermal effects of ultrasound in a nanodrug delivery system. 2022 IEEE International Ultrasonics Symposium (IUS), IEEE, 1–4.
  • Hornsby TK, Moradi Kashkooli F, Jakhmola A, et al. (2023). Multiphysics modeling of low-intensity pulsed ultrasound induced chemotherapeutic drug release from the surface of gold nanoparticles. Cancers 15:523.
  • Huber PE, Bischof M, Jenne J, et al. (2005). Trimodal cancer treatment: beneficial effects of combined antiangiogenesis, radiation, and chemotherapy. Cancer Res 65:3643–55.
  • Hynynen K. (2011). MRIgHIFU: a tool for image-guided therapeutics. J Magn Reson Imaging 34:482–93.
  • Islam MT, Tang S, Tasciotti E, Righetti R. (2021). Non-invasive assessment of the spatial and temporal distributions of interstitial fluid pressure, fluid velocity and fluid flow in cancers. Vivo, IEEE Access 9:89222–33. In PP
  • Izadifar Z, Babyn P, Chapman D. (2017). Mechanical and biological effects of ultrasound: a review of present knowledge. Ultrasound Med Biol 43:1085–104.
  • Jadidi A, Shokrgozar MA, Sardari S, Maadani AM. (2022). Gefitinib-loaded polydopamine-coated hollow mesoporous silica nanoparticle for gastric cancer application. Int J Pharm 629:122342.
  • Jain RK. (1987). Transport of molecules across tumor vasculature. Cancer Metastasis Rev 6:559–93.
  • Jain RK, Baxter LT. (1988). Mechanisms of heterogeneous distribution of monoclonal antibodies and other macromolecules in tumors: significance of elevated interstitial pressure. Cancer Res 48:7022–32.
  • Jain A, Tiwari A, Verma A, Jain SK. (2018). Ultrasound-based triggered drug delivery to tumors. Drug Deliv Transl Res 8:150–64.
  • Karimi M, Ghasemi A, Zangabad PS, et al. (2016). Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem Soc Rev 45:1457–501.
  • Kashkooli FM, Jakhmola A, Hornsby TK, et al. (2023). Ultrasound-mediated nano drug delivery for treating cancer: fundamental physics to future directions. J Control Release 355:552–78.
  • Kashkooli FM, Rezaeian M, Soltani M. (2022). Drug delivery through nanoparticles in solid tumors: a mechanistic understanding. Nanomedicine (Lond) 17:695–716.
  • Kashkooli FM, Soltani M, Rezaeian M, et al. (2020). Effect of vascular normalization on drug delivery to different stages of tumor progression: in-silico analysis. J Drug Delivery Sci Technol 60:101989.
  • Kashkooli FM, Soltani M, Souri M, et al. (2021). Nexus between in silico and in vivo models to enhance clinical translation of nanomedicine. Nano Today 36:101057.
  • Kashkooli FM, Soltani M, Souri M. (2020). Controlled anti-cancer drug release through advanced nano-drug delivery systems: static and dynamic targeting strategies. J Control Release 327:316–49.
  • Kim C, Guo Y, Velalopoulou A, et al. (2021). Closed-loop trans-skull ultrasound hyperthermia leads to improved drug delivery from thermosensitive drugs and promotes changes in vascular transport dynamics in brain tumors. Theranostics 11:7276–93.
  • Kneidl B, Peller M, Winter G, et al. (2014). Thermosensitive liposomal drug delivery systems: state of the art review. Int J Nanomedicine 9:4387–98.
  • Legha SS, Benjamin RS, Mackay B, et al. (1982). Reduction of doxorubicin cardiotoxicity by prolonged continuous intravenous infusion. Ann Intern Med 96:133–9.
  • Liu C, Xu XY. (2015). A systematic study of temperature sensitive liposomal delivery of doxorubicin using a mathematical model. Comput Biol Med 60:107–16.
  • Löke DR, Helderman RF, Franken NA, et al. (2021). Simulating drug penetration during hyperthermic intraperitoneal chemotherapy. Drug Deliv 28:145–61.
  • Lyon PC, Gray MD, Mannaris C, et al. (2018). Safety and feasibility of ultrasound-triggered targeted drug delivery of doxorubicin from thermosensitive liposomes in liver tumours (TARDOX): a single-centre, open-label, phase 1 trial. Lancet Oncol 19:1027–39.
  • Lyon PC, Gray M, Mannaris C, et al. (2019). Results of first-in-man proof of concept study of ultrasound-triggered drug delivery in liver tumours. Clin Radiol 74:e20.
  • Lyon PC, Griffiths LF, Lee J, et al. (2017). Clinical trial protocol for TARDOX: a phase I study to investigate the feasibility of targeted release of lyso-thermosensitive liposomal doxorubicin (ThermoDox®) using focused ultrasound in patients with liver tumours. J Ther Ultrasound 5:1–8.
  • Lyon PC, Mannaris C, Gray M, et al. (2021). Large-volume hyperthermia for safe and cost-effective targeted drug delivery using a clinical ultrasound-guided focused ultrasound device. Ultrasound Med Biol 47:982–97.
  • McDonald DM, Baluk P. (2002). Significance of blood vessel leakiness in cancer. Cancer Res 62:5381–5385.
  • Mi P. (2020). Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 10:4557–88.
  • Mihcin S, Melzer A. (2018). Principles of focused ultrasound. Minim Invasive Ther Allied Technol 27:41–50.
  • Moradi Kashkooli F, Soltani M. (2021). Evaluation of solid tumor response to sequential treatment cycles via a new computational hybrid approach. Sci Rep 11:1–15.
  • Moradi Kashkooli F, Soltani M, Momeni MM, Rahmim A. (2021). Enhanced drug delivery to solid tumors via drug-loaded nanocarriers: an image-based computational framework. Front Oncol 11:2252.
  • Mura S, Nicolas J, Couvreur P. (2013). Stimuli-responsive nanocarriers for drug delivery. Nat Mater 12:991–1003.
  • Namakshenas P, Mojra A. (2023). Efficient drug delivery to hypoxic tumors using thermosensitive liposomes with encapsulated anti-cancer drug under high intensity pulsed ultrasound. Int J Mech Sci 237:107818.
  • Needham D, Dewhirst MW. (2001). The development and testing of a new temperature-sensitive drug delivery system for the treatment of solid tumors. Adv Drug Deliv Rev 53:285–305.
  • Papaioannou L, Avgoustakis K. (2022). Responsive nanomedicines enhanced by or enhancing physical modalities to treat solid cancer tumors: preclinical and clinical evidence of safety and efficacy. Adv Drug Deliv Rev 181:114075.
  • Pishko GL, Astary GW, Mareci TH, Sarntinoranont M. (2011). Sensitivity analysis of an image-based solid tumor computational model with heterogeneous vasculature and porosity. Ann Biomed Eng 39:2360–73.
  • Rezaeian M, Sedaghatkish A, Soltani M. (2019). Numerical modeling of high-intensity focused ultrasound-mediated intraperitoneal delivery of thermosensitive liposomal doxorubicin for cancer chemotherapy. Drug Deliv 26:898–917.
  • Roohi R, Baroumand S, Hosseinie R, Ahmadi G. (2021). Numerical simulation of HIFU with dual transducers: the implementation of dual-phase lag bioheat and non-linear Westervelt equations. Int Commun Heat Mass Transfer 120:105002.
  • Sarmadi M, Ta C, VanLonkhuyzen AM, et al. (2022). Experimental and computational understanding of pulsatile release mechanism from biodegradable core-shell microparticles. Sci Adv 8:eabn5315.
  • Schultz CW, Ruiz de Garibay G, Langer A, et al. (2021). Selecting the optimal parameters for sonoporation of pancreatic cancer in a pre-clinical model. Cancer Biol Ther 22:204–15.
  • Schutt DJ, Haemmerich D. (2008). Effects of variation in perfusion rates and of perfusion models in computational models of radio frequency tumor ablation. Med Phys 35:3462–70.
  • Sedaghatkish A, Rezaeian M, Heydari H, et al. (2020). Acoustic streaming and thermosensitive liposomes for drug delivery into hepatocellular carcinoma tumor adjacent to major hepatic veins; an acoustics–thermal–fluid-mass transport coupling model. Int J Therm Sci 158:106540.
  • Seynhaeve A, Amin M, Haemmerich D, et al. (2020). Hyperthermia and smart drug delivery systems for solid tumor therapy. Adv Drug Delivery Rev 163-164:125–44.
  • Shaswary E, Assi H, Yang C, et al. (2021). Noninvasive calibrated tissue temperature estimation using backscattered energy of acoustic harmonics. Ultrasonics 114:106406.
  • Sheu TW, Solovchuk MA, Chen AW, Thiriet M. (2011). On an acoustics–thermal–fluid coupling model for the prediction of temperature elevation in liver tumor. Int J Heat Mass Transf 54:4117–26.
  • Singh G, Paul A, Shekhar H, Paul A. (2021). Pulsed ultrasound assisted thermo-therapy for subsurface tumor ablation: a numerical investigation. J Therm Sci Eng Appl 13:041007.
  • Sirsi SR, Borden MA. (2014). State-of-the-art materials for ultrasound-triggered drug delivery. Adv Drug Deliv Rev 72:3–14.
  • Solovchuk M, Sheu TW, Thiriet M. (2013). Simulation of nonlinear Westervelt equation for the investigation of acoustic streaming and nonlinear propagation effects. J Acoust Soc Am 134:3931–42.
  • Soltani M, Chen P. (2011). Numerical modeling of fluid flow in solid tumors. PloS One 6:e20344.
  • Soltani M,  Souri M, Moradi  Kashkooli F.  (2021). Effects  of hypoxia and nanocarrier size on pH-responsive nano-delivery  system  to solid tumors.  Sci  Rep 11:1–12.
  • Souri M, Moradi Kashkooli F, Soltani M. (2022). Analysis of magneto-hyperthermia duration in nano-sized drug delivery system to solid tumors using intravascular-triggered thermosensitive-liposome. Pharm Res 39:753–65.
  • Souri M, Soltani M, Kashkooli FM, et al. (2022). Towards principled design of cancer nanomedicine to accelerate clinical translation. Materials Today Bio 13:100208.
  • Souri M, Soltani M, Kashkooli FM, Shahvandi MK. (2022). Engineered strategies to enhance tumor penetration of drug-loaded nanoparticles. J Control Release 341:227–46.
  • Souri M, Soltani M, Moradi Kashkooli F. (2021). Computational modeling of thermal combination therapies by magneto-ultrasonic heating to enhance drug delivery to solid tumors. Sci Rep 11:1–12.
  • Staruch R, Chopra R, Hynynen K. (2011). Localised drug release using MRI-controlled focused ultrasound hyperthermia. Int J Hyperthermia 27:156–71.
  • Stylianopoulos T, Economides E-A, Baish JW, et al. (2015). Towards optimal design of cancer nanomedicines: multi-stage nanoparticles for the treatment of solid tumors. Ann Biomed Eng 43:2291–300.
  • Stylianopoulos T, Jain RK. (2013). Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc Natl Acad Sci U S A 110:18632–7.
  • Stylianopoulos T, Soteriou K, Fukumura D, Jain RK. (2013). Cationic nanoparticles have superior transvascular flux into solid tumors: insights from a mathematical model. Ann Biomed Eng 41:68–77.
  • Tagami T, Ernsting MJ, Li S-D. (2011). Optimization of a novel and improved thermosensitive liposome formulated with DPPC and a Brij surfactant using a robust in vitro system. J Control Release 154:290–7.
  • Tagami T, May JP, Ernsting MJ, Li S-D. (2012). A thermosensitive liposome prepared with a Cu2+ gradient demonstrates improved pharmacokinetics, drug delivery and antitumor efficacy. J Control Release 161:142–9.
  • Tan Q, Zou X, Ding Y, et al. (2018). The influence of dynamic tissue properties on HIFU hyperthermia: a numerical simulation study. Appl Sci 8:1933.
  • Ten Hagen TL, Dreher MR, Zalba S, et al. (2021). Drug transport kinetics of intravascular triggered drug delivery systems. Commun Biol 4:1–17.
  • Ter Haar G. (2016). HIFU tissue ablation: concept and devices. In: Escoffre JM, Bouakaz A, eds Therapeutic ultrasound. Advances in experimental medicine and biology. Vol 880. Cham: Springer 3–20.
  • Tharkar P, Varanasi R, Wong WSF, et al. (2019). Nano-enhanced drug delivery and therapeutic ultrasound for cancer treatment and beyond. Front Bioeng Biotechnol 7:324.
  • Ughachukwu P, Unekwe P, Pump E. (2012). Mediated resistance in chemotherapy. Ann Med Health Sci Res 2:191–8.
  • Van Durme R, Crevecoeur G, Dupré L, Coene A. (2021). Model-based optimized steering and focusing of local magnetic particle concentrations for targeted drug delivery. Drug Deliv 28:63–76.
  • Wang C-H, Li J, Teo CS, Lee T. (1999). The delivery of BCNU to brain tumors. J Control Release 61:21–41.
  • Zhan W. (2014). Mathematical modelling of drug delivery to solid tumour. PhD Thesis, Department of Chemical Engineering, Imperial College London, London, England.
  • Zhan W, Gedroyc W, Xu XY. (2019). Towards a multiphysics modelling framework for thermosensitive liposomal drug delivery to solid tumour combined with focused ultrasound hyperthermia. Biophys Rep 5:43–59.
  • Zhan W, Wang C-H. (2018). Convection enhanced delivery of liposome encapsulated doxorubicin for brain tumour therapy. J Control Release 285:212–29.
  • Zhang L, Lin Z, Zeng L, et al. (2022). Ultrasound-induced biophysical effects in controlled drug delivery. Sci China Life Sci 65:896–908.
  • Zhao J, Salmon H, Sarntinoranont M. (2007). Effect of heterogeneous vasculature on interstitial transport within a solid tumor. Microvasc Res 73:224–36.
  • Zou X, Dong H, Qian S-Y. (2020). Influence of dynamic tissue properties on temperature elevation and lesions during HIFU scanning therapy: numerical simulation. Chinese Phys B 29:034305.
  • Zou Y, Yamagishi M, Horikoshi I, et al. (1993). Enhanced therapeutic effect against liver W256 carcinosarcoma with temperature-sensitive liposomal adriamycin administered into the hepatic artery. Cancer Res 53:3046–51.