334
Views
9
CrossRef citations to date
0
Altmetric
Review

Mathematical modeling analysis of intratumoral disposition of anticancer agents and drug delivery systems

&

Bibliography

  • YHBae, KPark. Targeted drug delivery to tumors: myths, reality and possibility. J Control Release 2011;153:198–205
  • TLammers, WEHennink, GStorm. Tumour-targeted nanomedicines: principles and practice. Br J Cancer 2008;99:392–7
  • SDSteichen, MCaldorera-Moore, NAPeppas. A review of current nanoparticle and targeting moieties for the delivery of cancer therapeutics. Eur J Pharm Sci 2013;48:416–27
  • ABabu, AKTempleton, AMunshi, et al. Nanodrug delivery systems: a promising technology for detection, diagnosis, and treatment of cancer. AAPS PharmSciTech 2014;15:709–21
  • JWNichols, YHBae. Odyssey of a cancer nanoparticle: from injection site to site of action. Nano Today 2012;7:606–18
  • STaurin, HNehoff, KGreish. Anticancer nanomedicine and tumor vascular permeability; Where is the missing link? J Control Release 2012;164:265–75
  • AIMinchinton, IFTannock. Drug penetration in solid tumours. Nat Rev Cancer 2006;6:583–92
  • TLammers, FKiessling, WEHennink, et al. Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. J Control Release 2012;161:175–87
  • DStepensky. Pharmacokinetic and pharmacodynamic aspects of focal and targeted delivery of drugs. In: AJDomb, WKhan, editors. Focal controlled drug delivery. Springer US, New York; 2014
  • FDanhier, OFeron, VPreat. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010;148:135–46
  • HHolback, YYeo. Intratumoral drug delivery with nanoparticulate carriers. Pharm Res 2011;28:1819–30
  • YLi, JWang, MGWientjes, et al. Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor. Adv Drug Deliv Rev 2012;64:29–39
  • MJErnsting, MMurakami, ARoy, et al. Factors controlling the pharmacokinetics, biodistribution and intratumoral penetration of nanoparticles. J Control Release 2013;172:782–94
  • FMarcucci, ACorti. How to improve exposure of tumor cells to drugs - promoter drugs increase tumor uptake and penetration of effector drugs. Adv Drug Deliv Rev 2011;64:53–68
  • IAKhawar, JHKim, HKuh. Improving drug delivery to solid tumors: Priming the tumor microenvironment. J Control Release 2015;201C:78–89
  • MKanapathipillai, ABrock, DEIngber. Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment. Adv Drug Deliv Rev 2014;79-80:107–18
  • CWong, TStylianopoulos, JCui, et al. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc Natl Acad Sci USA 2011;108:2426–31
  • RESerda, BGodin, EBlanco, et al. Multi-stage delivery nano-particle systems for therapeutic applications. Biochim Biophys Acta 2011;1810:317–29
  • ASwierniak, MKimmel, JSmieja. Mathematical modeling as a tool for planning anticancer therapy. Eur J Pharmacol 2009;625:108–21
  • HMByrne. Dissecting cancer through mathematics: from the cell to the animal model. Nat Rev Cancer 2010;10:221–30
  • BDWeinberg, EBlanco, JGao. Polymer implants for intratumoral drug delivery and cancer therapy. J Pharm Sci 2008;97:1681–702
  • SMMoghimi, ACHunter, TLAndresen. Factors controlling nanoparticle pharmacokinetics: an integrated analysis and perspective. Annu Rev Pharmacol Toxicol 2011;52:481–503
  • ATFlorence. “Targeting” nanoparticles: the constraints of physical laws and physical barriers. J Control Release 2012;164:115–24
  • MKDanquah, XAZhang, RIMahato. Extravasation of polymeric nanomedicines across tumor vasculature. Adv Drug Deliv Rev 2011;63:623–39
  • LZhang, YWang, YCao, et al. Transport barriers and strategies of antitumor nanocarriers delivery system. J Biomed Mater Res A 2013;101:3661–9
  • HMaeda. Macromolecular therapeutics in cancer treatment: the EPR effect and beyond. J Control Release 2012;164:138–44
  • BRochat. Importance of influx and efflux systems and xenobiotic metabolizing enzymes in intratumoral disposition of anticancer agents. Curr Cancer Drug Targets 2009;9:652–74
  • Rvan der Meel, LJVehmeijer, RJKok, et al. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv Rev 2013;65:1284–98
  • FBouzom, KBall, NPerdaems, et al. Physiologically based pharmacokinetic (PBPK) modelling tools: how to fit with our needs? Biopharm Drug Dispos 2012;33:55–71
  • ESchuck, TBohnert, AChakravarty, et al. Preclinical pharmacokinetic/pharmacodynamic modeling and simulation in the pharmaceutical industry: an IQ consortium survey examining the current landscape. AAPS J 2015; In press
  • DEKirschner, JJLinderman. Mathematical and computational approaches can complement experimental studies of host-pathogen interactions. Cell Microbiol 2009;11:531–9
  • BDWeinberg, HAi, EBlanco, et al. Antitumor efficacy and local distribution of doxorubicin via intratumoral delivery from polymer millirods. J Biomed Mater Res A 2007;81:161–70
  • BDWeinberg, RBPatel, HWu, et al. Model simulation and experimental validation of intratumoral chemotherapy using multiple polymer implants. Med Biol Eng Comput 2008;46:1039–49
  • DYArifin, KYLee, CHWang. Chemotherapeutic drug transport to brain tumor. J Control Release 2009;137:203–10
  • DYArifin, KYLee, CHWang, et al. Role of convective flow in carmustine delivery to a brain tumor. Pharm Res 2009;26:2289–302
  • SHRanganath, YFu, DYArifin, et al. The use of submicron/nanoscale PLGA implants to deliver paclitaxel with enhanced pharmacokinetics and therapeutic efficacy in intracranial glioblastoma in mice. Biomaterials 2010;31:5199–207
  • AWadee, VPillay, YEChoonara, et al. Recent advances in the design of drug-loaded polymeric implants for the treatment of solid tumors. Expert Opin Drug Deliv 2011;8:1323–40
  • JBWolinsky, YLColson, MWGrinstaff. Local drug delivery strategies for cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers. J Control Release 2012;159:14–26
  • KCChen, JKim, XLi, et al. Modeling recombinant immunotoxin efficacies in solid tumors. Ann Biomed Eng 2008;36:486–512
  • RVenkatasubramanian, RBArenas, MAHenson, et al. Mechanistic modelling of dynamic MRI data predicts that tumour heterogeneity decreases therapeutic response. Br J Cancer 2010;103:486–97
  • MABattaglia, RSParker. Pharmacokinetic/pharmacodynamic modelling of intracellular gemcitabine triphosphate accumulation: translating in vitro to in vivo. IET Syst Biol 2011;5:34–43
  • MROwen, IJStamper, MMuthana, et al. Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy. Cancer Res 2011;71:2826–37
  • JLAu, PGuo, YGao, et al. Multiscale tumor spatiokinetic model for intraperitoneal therapy. AAPS J 2014;16:424–39
  • ESjogren, TLTammela, BLennernas, et al. Pharmacokinetics of an injectable modified-release 2-hydroxyflutamide formulation in the human prostate gland using a semiphysiologically based biopharmaceutical model. Mol Pharm 2014;11:3097–111
  • HDerendorf, BMeibohm. Modeling of pharmacokinetic/pharmacodynamic (PK/PD) relationships: concepts and perspectives. Pharm Res 1999;16:176–85
  • CEMeacham, SJMorrison. Tumour heterogeneity and cancer cell plasticity. Nature 2013;501:328–37
  • YBarenholz. Doxil(R) – the first FDA-approved nano-drug: lessons learned. J Control Release 2012;160:117–34
  • DHanahan, RAWeinberg. The hallmarks of cancer. Cell 2000;100:57–70
  • DHanahan, RAWeinberg. Hallmarks of cancer: the next generation. Cell 2011;144:646–74
  • SNegrini, VGGorgoulis, TDHalazonetis. Genomic instability – an evolving hallmark of cancer. Nat Rev Mol Cell Biol 2010;11:220–8
  • VFodale, MPierobon, LLiotta, et al. Mechanism of cell adaptation: when and how do cancer cells develop chemoresistance? Cancer J 2011;17:89–95
  • FQian, GMSaidel, DMSutton, et al. Combined modeling and experimental approach for the development of dual-release polymer millirods. J Control Release 2002;83:427–35
  • TTGoodman, JChen, KMatveev, et al. Spatio-temporal modeling of nanoparticle delivery to multicellular tumor spheroids. Biotechnol Bioeng 2008;101:388–99
  • WMok, TStylianopoulos, YBoucher, et al. Mathematical modeling of herpes simplex virus distribution in solid tumors: implications for cancer gene therapy. Clin Cancer Res 2009;15:2352–60
  • AZhang, XMi, GYang, et al. Numerical study of thermally targeted liposomal drug delivery in tumor. J Heat Transfer 2009;131:10.1115/1.3072952
  • MGWientjes, BZYeung, ZLu, et al. Predicting diffusive transport of cationic liposomes in 3-dimensional tumor spheroids. J Control Release 2014;192:10–18
  • MWu, HBFrieboes, MAChaplain, et al. The effect of interstitial pressure on therapeutic agent transport: coupling with the tumor blood and lymphatic vascular systems. J Theor Biol 2014;355:194–207
  • HHarashima, SIida, YUrakami, et al. Optimization of antitumor effect of liposomally encapsulated doxorubicin based on simulations by pharmacokinetic/pharmacodynamic modeling. J Control Release 1999;61:93–106
  • TLJackson, HMByrne. A mathematical model to study the effects of drug resistance and vasculature on the response of solid tumors to chemotherapy. Math Biosci 2000;164:17–38
  • TLJackson. Intracellular accumulation and mechanism of action of doxorubicin in a spatio-temporal tumor model. J Theor Biol 2003;220:201–13
  • MKohandel, MKardar, MMilosevic, et al. Dynamics of tumor growth and combination of anti-angiogenic and cytotoxic therapies. Phys Med Biol 2007;52:3665–77
  • RVenkatasubramanian, MAHenson, NSForbes. Integrating cell-cycle progression, drug penetration and energy metabolism to identify improved cancer therapeutic strategies. J Theor Biol 2008;253:98–117
  • SEikenberry. A tumor cord model for doxorubicin delivery and dose optimization in solid tumors. Theor Biol Med Model 2009;6:16
  • THara, SIriyama, KMakino, et al. Mathematical description of drug movement into tumor with EPR effect and estimation of its configuration for DDS. Colloids Surf B Biointerfaces 2010;75:42–6
  • MKohandel, CAHaselwandter, MKardar, et al. Quantitative model for efficient temporal targeting of tumor cells and neovasculature. Comput Math Methods Med 2011;2011:790721
  • GMThurber, RWeissleder. A systems approach for tumor pharmacokinetics. PLoS One 2011;6:e24696
  • AGasselhuber, MRDreher, FRattay, et al. Comparison of conventional chemotherapy, stealth liposomes and temperature-sensitive liposomes in a mathematical model. PLoS One 2012;7:e47453
  • JJRhoden, KDWittrup. Dose dependence of intratumoral perivascular distribution of monoclonal antibodies. J Pharm Sci 2012;101:860–7
  • DKShah, NHaddish-Berhane, ABetts. Bench to bedside translation of antibody drug conjugates using a multiscale mechanistic PK/PD model: a case study with brentuximab-vedotin. J Pharmacokinet Pharmacodyn 2012;39:643–59
  • ALvan de Ven, MWu, JLowengrub, et al. Integrated intravital microscopy and mathematical modeling to optimize nanotherapeutics delivery to tumors. AIP Adv 2012;2:11208
  • HBFrieboes, MWu, JLowengrub, et al. A computational model for predicting nanoparticle accumulation in tumor vasculature. PLoS One 2013;8:e56876
  • JSharma, HLv, JMGallo. Intratumoral modeling of gefitinib pharmacokinetics and pharmacodynamics in an orthotopic mouse model of glioblastoma. Cancer Res 2013;73:5242–52
  • WZhan, XYXu. A mathematical model for thermosensitive liposomal delivery of doxorubicin to solid tumour. J Drug Deliv 2013;2013:172529
  • JFStrasser, LKFung, SEller, et al. Distribution of 1,3-bis(2-chloroethyl)-1-nitrosourea and tracers in the rabbit brain after interstitial delivery by biodegradable polymer implants. J Pharmacol Exp Ther 1995;275:1647–55
  • LKFung, MShin, BTyler, et al. Chemotherapeutic drugs released from polymers: distribution of 1,3-bis(2-chloroethyl)-1-nitrosourea in the rat brain. Pharm Res 1996;13:671–82
  • LKFung, MGEwend, ASills, et al. Pharmacokinetics of interstitial delivery of carmustine, 4-hydroperoxycyclophosphamide, and paclitaxel from a biodegradable polymer implant in the monkey brain. Cancer Res 1998;58:672–84
  • RPort, FHanisch, MBecker, et al. Local disposition kinetics of floxuridine after intratumoral and subcutaneous injection as monitored by [19F]-nuclear magnetic resonance spectroscopy in vivo. Cancer Chemother Pharmacol 1999;44:65–73
  • ABFleming, WMSaltzman. Pharmacokinetics of the carmustine implant. Clin Pharmacokinet 2002;41:403–19
  • WHTan, TLee, CHWang. Simulation of intratumoral release of etanidazole: effects of the size of surgical opening. J Pharm Sci 2003;92:773–89
  • WHTan, FWang, TLee, et al. Computer simulation of the delivery of etanidazole to brain tumor from PLGA wafers: comparison between linear and double burst release systems. Biotechnol Bioeng 2003;82:278–88
  • ARTzafriri, EILerner, MFlashner-Barak, et al. Mathematical modeling and optimization of drug delivery from intratumorally injected microspheres. Clin Cancer Res 2005;11:826–34
  • DDelli Castelli, WDastru, ETerreno, et al. In vivo MRI multicontrast kinetic analysis of the uptake and intracellular trafficking of paramagnetically labeled liposomes. J Control Release 2010;144:271–9
  • AJTorres, CZhu, MLShuler, et al. Paclitaxel delivery to brain tumors from hydrogels: a computational study. Biotechnol Prog 2011;27:1478–87

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.