51
|
Magnetic resonance guided high-intensity focused ultrasound for image-guided temperature-induced drug delivery. Adv Drug Deliv Rev 2014; 72:65-81. [PMID: 24463345 DOI: 10.1016/j.addr.2014.01.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/08/2014] [Accepted: 01/14/2014] [Indexed: 12/29/2022]
Abstract
Magnetic resonance guided high-intensity focused ultrasound (MR-HIFU) is a versatile technology platform for noninvasive thermal therapies in oncology. Since MR-HIFU allows heating of deep-seated tissue to well-defined temperatures under MR image guidance, this novel technology has great potential for local heat-mediated drug delivery from temperature-sensitive liposomes (TSLs). In particular, MR provides the ability for image guidance of the drug delivery when an MRI contrast agent is co-encapsulated with the drug in the aqueous lumen of the liposomes. Monitoring of the tumor drug coverage offers possibilities for a personalized thermal treatment in oncology. This review focuses on MR-HIFU as a noninvasive technology platform, temperature-sensitive liposomal formulations for drug delivery and image-guided drug delivery, and the effect of HIFU-induced hyperthermia on the TSL and drug distribution. Finally, the opportunities and challenges of localized MR-HIFU-mediated drug delivery from temperature-sensitive liposomes in oncology are discussed.
Collapse
|
52
|
Therapeutic efficacy of combining pegylated liposomal doxorubicin and radiofrequency (RF) ablation: comparison between slow-drug-releasing, non-thermosensitive and fast-drug-releasing, thermosensitive nano-liposomes. PLoS One 2014; 9:e92555. [PMID: 24786533 PMCID: PMC4006748 DOI: 10.1371/journal.pone.0092555] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 02/25/2014] [Indexed: 11/19/2022] Open
Abstract
AIMS To determine how the accumulation of drug in mice bearing an extra-hepatic tumor and its therapeutic efficacy are affected by the type of PEGylated liposomal doxorubicin used, treatment modality, and rate of drug release from the liposomes, when combined with radiofrequency (RF) ablation. MATERIALS AND METHODS Two nano-drugs, both long-circulating PEGylated doxorubicin liposomes, were formulated: (1) PEGylated doxorubicin in thermosensitive liposomes (PLDTS), having a burst-type fast drug release above the liposomes' solid ordered to liquid disordered phase transition (at 42°C), and (2) non-thermosensitive PEGylated doxorubicin liposomes (PLDs), having a slow and continuous drug release. Both were administered intravenously at 8 mg/kg doxorubicin dose to tumor-bearing mice. Animals were divided into 6 groups: no treatment, PLD, RF, RF+PLD, PLDTS, and PLDTS+RF, for intra-tumor doxorubicin deposition at 1, 24, and 72 h post-injection (in total 41, mice), and 31 mice were used for randomized survival studies. RESULTS Non-thermosensitive PLD combined with RF had the least tumor growth and the best end-point survival, better than PLDTS+RF (p<0.005) or all individual therapies (p<0.001). Although at 1 h post-treatment the greatest amount of intra-tumoral doxorubicin was seen following PLDTS+RF (p<0.05), by 24 and 72 h the greatest doxorubicin amount was seen for PLD+RF (p<0.05); in this group the tumor also has the longest exposure to doxorubicin. CONCLUSION Optimizing therapeutic efficacy of PLD requires a better understanding of the relationship between the effect of RF on tumor microenvironment and liposome drug release profile. If drug release is too fast, the benefit of changing the microenvironment by RF on tumor drug localization and therapeutic efficacy may be much smaller than for PLDs having slow and temperature-independent drug release. Thus the much longer circulation time of doxorubicin from PLD than from PLDTS may be beneficial in many therapeutic instances, especially in extra-hepatic tumors.
Collapse
|
53
|
Tejera-Garcia R, Parkkila P, Zamotin V, Kinnunen PKJ. Principles of rational design of thermally targeted liposomes for local drug delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1243-52. [PMID: 24685945 DOI: 10.1016/j.nano.2014.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/10/2014] [Accepted: 03/20/2014] [Indexed: 02/01/2023]
Abstract
UNLABELLED Drug release from 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) liposomes occurs close to the main transition temperature Tm=41°C. The exact release temperature can be adjusted by additional lipids, which shift Tm. A major issue is drug leakage at 37°C. We here describe a novel approach with improved drug retention yet rapid release. To obtain spherical, smooth liposomes we included: i) 2mol% cholesterol, to soften bilayers (Lemmich et al 1997), ii) lipids, which due to their spontaneous curvature stabilize the negative and positive curvatures of the inner and outer leaflets of unilamellar liposomes. In addition to differential scanning calorimetry (DSC) and fluorescence spectroscopy, the lipid mixtures were analyzed by a Langmuir balance for their elastic properties and lipid packing, aiming at high elasticity modulus CS(-1). Maxima in CS(-1) coincided with minima in the free energy of lateral mixing. These liposomes have reduced drug leakage, yet retain rapid release. FROM THE CLINICAL EDITOR This paper reports the development of optimized DPPC liposomes for drug delivery, with reduced drug leakage but maintained rapid release.
Collapse
Affiliation(s)
- Roberto Tejera-Garcia
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, School of Sciences, Aalto University, Espoo, Finland
| | - Petteri Parkkila
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, School of Sciences, Aalto University, Espoo, Finland
| | - Vladimir Zamotin
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, School of Sciences, Aalto University, Espoo, Finland
| | - Paavo K J Kinnunen
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, School of Sciences, Aalto University, Espoo, Finland.
| |
Collapse
|
54
|
Affiliation(s)
- Chun Li
- Department of Cancer Systems Imaging—Unit 59, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas 77030, Tel: 713-792-5182,
| |
Collapse
|
55
|
Rossmann C, Haemmerich D. Review of temperature dependence of thermal properties, dielectric properties, and perfusion of biological tissues at hyperthermic and ablation temperatures. Crit Rev Biomed Eng 2014; 42:467-92. [PMID: 25955712 PMCID: PMC4859435 DOI: 10.1615/critrevbiomedeng.2015012486] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The application of supraphysiological temperatures (>40°C) to biological tissues causes changes at the molecular, cellular, and structural level, with corresponding changes in tissue function and in thermal, mechanical and dielectric tissue properties. This is particularly relevant for image-guided thermal treatments (e.g. hyperthermia and thermal ablation) delivering heat via focused ultrasound (FUS), radiofrequency (RF), microwave (MW), or laser energy; temperature induced changes in tissue properties are of relevance in relation to predicting tissue temperature profile, monitoring during treatment, and evaluation of treatment results. This paper presents a literature survey of temperature dependence of electrical (electrical conductivity, resistivity, permittivity) and thermal tissue properties (thermal conductivity, specific heat, diffusivity). Data of soft tissues (liver, prostate, muscle, kidney, uterus, collagen, myocardium and spleen) for temperatures between 5 to 90°C, and dielectric properties in the frequency range between 460 kHz and 3 GHz are reported. Furthermore, perfusion changes in tumors including carcinomas, sarcomas, rhabdomyosarcoma, adenocarcinoma and ependymoblastoma in response to hyperthmic temperatures up to 46°C are presented. Where appropriate, mathematical models to describe temperature dependence of properties are presented. The presented data is valuable for mathematical models that predict tissue temperature during thermal therapies (e.g. hyperthermia or thermal ablation), as well as for applications related to prediction and monitoring of temperature induced tissue changes.
Collapse
Affiliation(s)
- Christian Rossmann
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA; Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
56
|
Stapleton S, Milosevic M, Allen C, Zheng J, Dunne M, Yeung I, Jaffray DA. A mathematical model of the enhanced permeability and retention effect for liposome transport in solid tumors. PLoS One 2013; 8:e81157. [PMID: 24312530 PMCID: PMC3846845 DOI: 10.1371/journal.pone.0081157] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 10/09/2013] [Indexed: 11/25/2022] Open
Abstract
The discovery of the enhanced permeability and retention (EPR) effect has resulted in the development of nanomedicines, including liposome-based formulations of drugs, as cancer therapies. The use of liposomes has resulted in substantial increases in accumulation of drugs in solid tumors; yet, significant improvements in therapeutic efficacy have yet to be achieved. Imaging of the tumor accumulation of liposomes has revealed that this poor or variable performance is in part due to heterogeneous inter-subject and intra-tumoral liposome accumulation, which occurs as a result of an abnormal transport microenvironment. A mathematical model that relates liposome accumulation to the underlying transport properties in solid tumors could provide insight into inter and intra-tumoral variations in the EPR effect. In this paper, we present a theoretical framework to describe liposome transport in solid tumors. The mathematical model is based on biophysical transport equations that describe pressure driven fluid flow across blood vessels and through the tumor interstitium. The model was validated by direct comparison with computed tomography measurements of tumor accumulation of liposomes in three preclinical tumor models. The mathematical model was fit to liposome accumulation curves producing predictions of transport parameters that reflect the tumor microenvironment. Notably, all fits had a high coefficient of determination and predictions of interstitial fluid pressure agreed with previously published independent measurements made in the same tumor type. Furthermore, it was demonstrated that the model attributed inter-subject heterogeneity in liposome accumulation to variations in peak interstitial fluid pressure. These findings highlight the relationship between transvascular and interstitial flow dynamics and variations in the EPR effect. In conclusion, we have presented a theoretical framework that predicts inter-subject and intra-tumoral variations in the EPR effect based on fundamental properties of the tumor microenvironment and forms the basis for transport modeling of liposome drug delivery.
Collapse
Affiliation(s)
- Shawn Stapleton
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- * E-mail:
| | - Michael Milosevic
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jinzi Zheng
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Michael Dunne
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ivan Yeung
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - David A. Jaffray
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
- STTARR Innovation Centre, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Techna Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
57
|
Kim M, Gillies RJ, Rejniak KA. Current advances in mathematical modeling of anti-cancer drug penetration into tumor tissues. Front Oncol 2013; 3:278. [PMID: 24303366 PMCID: PMC3831268 DOI: 10.3389/fonc.2013.00278] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/29/2013] [Indexed: 11/26/2022] Open
Abstract
Delivery of anti-cancer drugs to tumor tissues, including their interstitial transport and cellular uptake, is a complex process involving various biochemical, mechanical, and biophysical factors. Mathematical modeling provides a means through which to understand this complexity better, as well as to examine interactions between contributing components in a systematic way via computational simulations and quantitative analyses. In this review, we present the current state of mathematical modeling approaches that address phenomena related to drug delivery. We describe how various types of models were used to predict spatio-temporal distributions of drugs within the tumor tissue, to simulate different ways to overcome barriers to drug transport, or to optimize treatment schedules. Finally, we discuss how integration of mathematical modeling with experimental or clinical data can provide better tools to understand the drug delivery process, in particular to examine the specific tissue- or compound-related factors that limit drug penetration through tumors. Such tools will be important in designing new chemotherapy targets and optimal treatment strategies, as well as in developing non-invasive diagnosis to monitor treatment response and detect tumor recurrence.
Collapse
Affiliation(s)
- Munju Kim
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL , USA
| | | | | |
Collapse
|
58
|
Hong CW, Libutti SK, Wood BJ. Liposomal doxorubicin plus radiofrequency ablation for complete necrosis of a hepatocellular carcinoma. ACTA ACUST UNITED AC 2013; 20:e274-7. [PMID: 23737698 DOI: 10.3747/co.20.1266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Radiofrequency ablation (rfa) is a standard treatment for small, unresectable hepatocellular carcinomas (hccs). However, rfa for larger tumours is less successful, and intravenous lyso-thermosensitive liposomal doxorubicin during rfa is one technique postulated to potentially address that limitation. This drug-plus-device combination therapy was used to completely treat a hcc in a patient who underwent liver transplantation 79 days later.
Collapse
Affiliation(s)
- C W Hong
- Center for Interventional Oncology, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD, U.S.A. ; Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, U.S.A
| | | | | |
Collapse
|
59
|
Staruch RM, Ganguly M, Tannock IF, Hynynen K, Chopra R. Enhanced drug delivery in rabbit VX2 tumours using thermosensitive liposomes and MRI-controlled focused ultrasound hyperthermia. Int J Hyperthermia 2013; 28:776-87. [PMID: 23153219 DOI: 10.3109/02656736.2012.736670] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE The efficacy of anticancer drugs in solid tumours is impaired by their inability to reach all cancer cells in sufficient concentration to cause cytotoxicity. Hyperthermia-triggered release of drugs from thermosensitive liposomes can increase tumour drug concentration, but tumour-specific drug delivery requires precise temperature control, and effects on microregional distribution of anticancer drugs in tumours are unknown. Here we evaluate thermally triggered release of doxorubicin in a rabbit tumour model by comparing free versus thermosensitive liposomal doxorubicin administered systemically during magnetic resonance imaging (MRI)-controlled focused ultrasound hyperthermia. MATERIALS AND METHODS Twelve rabbits with a transplanted VX2 tumour in each thigh had a 10 mm diameter region in one tumour heated to 43°C using focused ultrasound with temperature control by MRI thermometry. Delivery of doxorubicin to tumours and normal tissues was quantified by fluorescence in tissue homogenates, and by fluorescence microscopy. RESULTS Using thermosensitive liposomal doxorubicin (2.5 mg/kg), doxorubicin concentrations in heated tumours were 26.7 times higher than in unheated tumours (n = 7, p = 0.017, two-sided Wilcoxon signed-rank test). There was no significant enhancement with free doxorubicin in heated versus unheated tumours (n = 3, p = 0.5). With thermosensitive liposomes (8.3 mg/kg), fluorescence microscopy demonstrated increased doxorubicin fluorescence in heated versus unheated tumours, co-localised with nuclear staining throughout the tumour. CONCLUSIONS Localised image-guided delivery of high concentrations of doxorubicin to cancer cells was achieved non-invasively in implanted tumours with temperature-sensitive drug carriers and a preclinical MRI-controlled focused ultrasound hyperthermia system.
Collapse
Affiliation(s)
- Robert M Staruch
- Centre for Research in Image-Guided Therapeutics, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
60
|
Li L, ten Hagen TLM, Hossann M, Süss R, van Rhoon GC, Eggermont AMM, Haemmerich D, Koning GA. Mild hyperthermia triggered doxorubicin release from optimized stealth thermosensitive liposomes improves intratumoral drug delivery and efficacy. J Control Release 2013; 168:142-50. [PMID: 23524188 DOI: 10.1016/j.jconrel.2013.03.011] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 03/09/2013] [Accepted: 03/15/2013] [Indexed: 12/15/2022]
Abstract
Liposome mediated anticancer drug delivery has the advantage of reducing cytotoxicity in healthy tissues. However, undesired slow drug release impedes the therapeutic efficacy of clinically applied PEG-liposomal doxorubicin (Dox). The aim of this study is to combine stealth thermosensitive liposomes (TSL) and local mild hyperthermia (HT) to increase bioavailable Dox levels in tumors. Dox was encapsulated in stealth TSL (~80nm) with optimized PEG concentration in the membrane, and compared with lysolipid-based Dox-LTSL for in vitro stability, release kinetics, and in vivo tumor growth control. In vitro cytotoxicity of Dox-TSL against murine BFS-1 sarcoma and, human BLM melanoma cell lines and Human Umbilical Vein Endothelial Cells (HUVEC) under normothermia (37°C) and HT (42°C) was compared with non-encapsulated Dox. In vitro Dox uptake in nuclei was imaged in BLM and HUVEC. In vivo intravascular Dox release from TSL in BFS-1 tumors under local mild HT in dorsal skin flap window chamber models was captured by intravital confocal microscopy. Intravascular Dox-TSL release kinetics, penetration depth and interstitial Dox density were subjected to quantitative image analysis. Systemic Dox-TSL administration in combination with local mild HT on subcutaneous tumor growth control was compared to Dox-LTSL plus local mild HT. Dox-TSL was stable at 37°C, while released over 95% Dox within 1min in 90% serum at 42°C. Dox-TSL demonstrated efficient in vivo intratumoral Dox release under local mild HT, followed by significant Dox uptake by tumor and tumor vascular endothelial cells. Dox-TSL plus mild HT showed improved tumor growth control over Dox-LTSL plus mild HT. Survival after a single treatment of Dox-TSL plus mild HT was 67%, while survival after Dox-LTSL plus mild HT was 22%. This combination of Dox-TSL and local mild HT offers promising clinical opportunities to improve liposomal Dox delivery to solid tumors.
Collapse
Affiliation(s)
- Li Li
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Dewhirst MW, Landon CD, Hofmann CL, Stauffer PR. Novel approaches to treatment of hepatocellular carcinoma and hepatic metastases using thermal ablation and thermosensitive liposomes. Surg Oncol Clin N Am 2013; 22:545-61. [PMID: 23622079 DOI: 10.1016/j.soc.2013.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Because of the limitations of surgical resection, thermal ablation is commonly used for the treatment of hepatocellular carcinoma and liver metastases. Current methods of ablation can result in marginal recurrences of larger lesions and in tumors located near large vessels. This review presents a novel approach for extending treatment out to the margins where temperatures do not provide complete treatment with ablation alone, by combining thermal ablation with drug-loaded thermosensitive liposomes. A history of the development of thermosensitive liposomes is presented. Clinical trials have shown that the combination of radiofrequency ablation and doxorubicin-loaded thermosensitive liposomes is a promising treatment.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Radiation Oncology Department, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | |
Collapse
|
62
|
Cervadoro A, Giverso C, Pande R, Sarangi S, Preziosi L, Wosik J, Brazdeikis A, Decuzzi P. Design maps for the hyperthermic treatment of tumors with superparamagnetic nanoparticles. PLoS One 2013; 8:e57332. [PMID: 23451208 PMCID: PMC3581487 DOI: 10.1371/journal.pone.0057332] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 01/18/2013] [Indexed: 11/19/2022] Open
Abstract
A plethora of magnetic nanoparticles has been developed and investigated under different alternating magnetic fields (AMF) for the hyperthermic treatment of malignant tissues. Yet, clinical applications of magnetic hyperthermia are sporadic, mostly due to the low energy conversion efficiency of the metallic nanoparticles and the high tissue concentrations required. Here, we study the hyperthermic performance of commercially available formulations of superparamagnetic iron oxide nanoparticles (SPIOs), with core diameter of 5, 7 and 14 nm, in terms of absolute temperature increase ΔT and specific absorption rate (SAR). These nanoparticles are operated under a broad range of AMF conditions, with frequency f varying between 0.2 and 30 MHz; field strength H ranging from 4 to 10 kA m(-1); and concentration cMNP varying from 0.02 to 3.5 mg ml(-1). At high frequency field (∼30 MHz), non specific heating dominates and ΔT correlates with the electrical conductivity of the medium. At low frequency field (<1 MHz), non specific heating is negligible and the relaxation of the SPIO within the AMF is the sole energy source. We show that the ΔT of the medium grows linearly with cMNP , whereas the SARMNP of the magnetic nanoparticles is independent of cMNP and varies linearly with f and H(2) . Using a computational model for heat transport in a biological tissue, the minimum requirements for local hyperthermia (Ttissue >42°C) and thermal ablation (Ttissue >50°C) are derived in terms of cMNP , operating AMF conditions and blood perfusion. The resulting maps can be used to rationally design hyperthermic treatments and identifying the proper route of administration - systemic versus intratumor injection - depending on the magnetic and biodistribution properties of the nanoparticles.
Collapse
Affiliation(s)
- Antonio Cervadoro
- Department of Translational Imaging, The Methodist Hospital Research Institute, Houston, Texas, United States of America
- Department of Mechanics, Politecnico di Torino, Turin, Italy
| | - Chiara Giverso
- Department of Mathematical Sciences, Politecnico di Torino, Turin, Italy
| | - Rohit Pande
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, United States of America
- Texas Superconductivity Center, Houston, Texas, United States of America
| | - Subhasis Sarangi
- Texas Superconductivity Center, Houston, Texas, United States of America
| | - Luigi Preziosi
- Department of Mathematical Sciences, Politecnico di Torino, Turin, Italy
| | - Jarek Wosik
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, United States of America
- Texas Superconductivity Center, Houston, Texas, United States of America
| | - Audrius Brazdeikis
- Texas Superconductivity Center, Houston, Texas, United States of America
- Department of Physics, University of Houston, Houston, Texas, United States of America
| | - Paolo Decuzzi
- Department of Translational Imaging, The Methodist Hospital Research Institute, Houston, Texas, United States of America
- Department of Experimental and Clinical Medicine, University of “Magna Graecia”, Catanzaro, Italy
| |
Collapse
|
63
|
Al-Ahmady ZS, Al-Jamal WT, Bossche JV, Bui TT, Drake AF, Mason AJ, Kostarelos K. Lipid-peptide vesicle nanoscale hybrids for triggered drug release by mild hyperthermia in vitro and in vivo. ACS NANO 2012; 6:9335-46. [PMID: 22857653 PMCID: PMC3480335 DOI: 10.1021/nn302148p] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The present study describes leucine zipper peptide-lipid hybrid nanoscale vesicles engineered by self-assembled anchoring of the amphiphilic peptide within the lipid bilayer. These hybrid vesicles aim to combine the advantages of traditional temperature-sensitive liposomes (TSL) with the dissociative, unfolding properties of a temperature-sensitive peptide to optimize drug release under mild hyperthermia, while improving in vivo drug retention. The secondary structure of the peptide and its thermal responsiveness after anchoring onto liposomes were studied with circular dichroism. In addition, the lipid-peptide vesicles (Lp-peptide) showed a reduction in bilayer fluidity at the inner core, as observed with DPH anisotropy studies, while the opposite effect was observed with an ANS probe, indicating peptide interactions with both the headgroup region and the hydrophobic core. A model drug molecule, doxorubicin, was successfully encapsulated in the Lp-peptide vesicles at higher than 90% efficiency following the remote loading, pH-gradient methodology. The release of doxorubicin from Lp-peptide hybrids in vitro indicated superior serum stability at physiological temperatures compared to lysolipid-containing temperature-sensitive liposomes (LTSL) without affecting the overall thermo-responsive nature of the vesicles at 42 °C. A similar stabilizing effect was observed in vivo after intravenous administration of the Lp-peptide vesicles by measuring (14)C-doxorubicin blood kinetics that also led to increased tumor accumulation after 24 h. We conclude that Lp-peptide hybrid vesicles present a promising new class of TSL that can offer previously unexplored opportunities for the development of clinically relevant mild hyperthermia-triggered therapeutic modalities.
Collapse
Affiliation(s)
- Zahraa S. Al-Ahmady
- Nanomedicine Lab, Centre for Drug Delivery Research, UCL School of Pharmacy, University College London, WC1N 1AX, United Kingdom
| | - Wafa’ T. Al-Jamal
- Nanomedicine Lab, Centre for Drug Delivery Research, UCL School of Pharmacy, University College London, WC1N 1AX, United Kingdom
| | - Jeroen V. Bossche
- Nanomedicine Lab, Centre for Drug Delivery Research, UCL School of Pharmacy, University College London, WC1N 1AX, United Kingdom
| | - Tam T. Bui
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Alex F. Drake
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - A. James Mason
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Centre for Drug Delivery Research, UCL School of Pharmacy, University College London, WC1N 1AX, United Kingdom
| |
Collapse
|
64
|
Comparison of conventional chemotherapy, stealth liposomes and temperature-sensitive liposomes in a mathematical model. PLoS One 2012; 7:e47453. [PMID: 23082168 PMCID: PMC3474827 DOI: 10.1371/journal.pone.0047453] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 09/17/2012] [Indexed: 12/26/2022] Open
Abstract
Various liposomal drug carriers have been developed to overcome short plasma half-life and toxicity related side effects of chemotherapeutic agents. We developed a mathematical model to compare different liposome formulations of doxorubicin (DOX): conventional chemotherapy (Free-DOX), Stealth liposomes (Stealth-DOX), temperature sensitive liposomes (TSL) with intra-vascular triggered release (TSL-i), and TSL with extra-vascular triggered release (TSL-e). All formulations were administered as bolus at a dose of 9 mg/kg. For TSL, we assumed locally triggered release due to hyperthermia for 30 min. Drug concentrations were determined in systemic plasma, aggregate body tissue, cardiac tissue, tumor plasma, tumor interstitial space, and tumor cells. All compartments were assumed perfectly mixed, and represented by ordinary differential equations. Contribution of liposomal extravasation was negligible in the case of TSL-i, but was the major delivery mechanism for Stealth-DOX and for TSL-e. The dominant delivery mechanism for TSL-i was release within the tumor plasma compartment with subsequent tissue- and cell uptake of released DOX. Maximum intracellular tumor drug concentrations for Free-DOX, Stealth-DOX, TSL-i, and TSL-e were 3.4, 0.4, 100.6, and 15.9 µg/g, respectively. TSL-i and TSL-e allowed for high local tumor drug concentrations with reduced systemic exposure compared to Free-DOX. While Stealth-DOX resulted in high tumor tissue concentrations compared to Free-DOX, only a small fraction was bioavailable, resulting in little cellular uptake. Consistent with clinical data, Stealth-DOX resulted in similar tumor intracellular concentrations as Free-DOX, but with reduced systemic exposure. Optimal release time constants for maximum cellular uptake for Stealth-DOX, TSL-e, and TSL-i were 45 min, 11 min, and <3 s, respectively. Optimal release time constants were shorter for MDR cells, with ∼4 min for Stealth-DOX and for TSL-e. Tissue concentrations correlated well quantitatively with a prior in-vivo study. Mathematical models may thus allow optimization of drug delivery systems to achieve a better therapeutic index.
Collapse
|
65
|
Gasselhuber A, Dreher MR, Partanen A, Yarmolenko PS, Woods D, Wood BJ, Haemmerich D. Targeted drug delivery by high intensity focused ultrasound mediated hyperthermia combined with temperature-sensitive liposomes: computational modelling and preliminary in vivovalidation. Int J Hyperthermia 2012; 28:337-48. [PMID: 22621735 DOI: 10.3109/02656736.2012.677930] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE To develop and validate a computational model that simulates (1) tissue heating with high intensity focused ultrasound (HIFU), and (2) resulting hyperthermia-mediated drug delivery from temperature-sensitive liposomes (TSL). MATERIALS AND METHODS HIFU heating in tissue was simulated using a heat transfer model based on the bioheat equation, including heat-induced cessation of perfusion. A spatio-temporal multi-compartment pharmacokinetic model simulated intravascular release of doxorubicin from TSL, its transport into interstitium, and cell uptake. Two heating schedules were simulated, each lasting 30 min: (1) hyperthermia at 43 °C (HT) and (2) hyperthermia followed by a high temperature (50 °C for 20 s) pulse (HT+). As preliminary model validation, in vivo studies were performed in thigh muscle of a New Zealand White rabbit, where local hyperthermia with a clinical magnetic resonance-guided HIFU system was applied following TSL administration. RESULTS HT produced a defined region of high doxorubicin concentration (cellular concentration ∼15-23 µg/g) in the target region. Cellular drug uptake was directly related to HT duration, with increasing doxorubicin uptake up to ∼2 h. HT+ enhanced drug delivery by ∼40% compared to HT alone. Temperature difference between model and experiment within the hyperthermia zone was on average 0.54 °C. Doxorubicin concentration profile agreed qualitatively with in vivo fluorescence profile. CONCLUSIONS Computational models can predict temperature and delivered drug from combination of HIFU with TSL. Drug delivery using TSL may be enhanced by prolonged hyperthermia up to 2 h or by local cessation of vascular perfusion with a high temperature pulse following hyperthermia.
Collapse
Affiliation(s)
- Astrid Gasselhuber
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | |
Collapse
|
66
|
Grüll H, Langereis S. Hyperthermia-triggered drug delivery from temperature-sensitive liposomes using MRI-guided high intensity focused ultrasound. J Control Release 2012; 161:317-27. [PMID: 22565055 DOI: 10.1016/j.jconrel.2012.04.041] [Citation(s) in RCA: 247] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/18/2012] [Accepted: 04/27/2012] [Indexed: 01/16/2023]
Abstract
In the continuous search for cancer therapies with a higher therapeutic window, localized temperature-induced drug delivery may offer a minimal invasive treatment option. Here, a chemotherapeutic drug is encapsulated into a temperature-sensitive liposome (TSL) that is released at elevated temperatures, for example, when passing through a locally heated tumor. Consequently, high drug levels in the tumor tissue can be achieved, while reducing drug exposure to healthy tissue. Although the concept of temperature-triggered drug delivery was suggested more than thirty years ago, several chemical and technological challenges had to be addressed to advance this approach towards clinical translation. In particular, non-invasive focal heating of tissue in a controlled fashion remained a challenge. For the latter, high intensity focused ultrasound (HIFU) allows non-invasive heating to establish hyperthermia (40-45 °C) of tumor tissue over time. Magnetic resonance imaging (MRI) plays a pivotal role in this procedure thanks to its superb spatial resolution for soft tissue as well as the possibility to acquire 3D temperature information. Consequently, MRI systems emerged with an HIFU ultrasound transducer embedded in the patient bed (MR-HIFU), where the MRI is utilized for treatment planning, and to provide spatial and temperature feedback to the HIFU. For tumor treatment, the lesion is heated to 42 °C using HIFU. At this temperature, the drug-loaded TSLs release their payload in a quantitative fashion. The concept of temperature-triggered drug delivery has been extended to MR image-guided drug delivery by the co-encapsulation of a paramagnetic MRI contrast agent in the lumen of TSLs. This review will give an overview of recent developments in temperature-induced drug delivery using HIFU under MRI guidance.
Collapse
Affiliation(s)
- Holger Grüll
- Eindhoven University of Technology, Department of Biomedical NMR, Eindhoven, The Netherlands.
| | | |
Collapse
|
67
|
Ranjan A, Jacobs G, Woods DL, Negussie AH, Partanen A, Yarmolenko PS, Gacchina CE, Sharma KV, Frenkel V, Wood BJ, Dreher MR. Image-guided drug delivery with magnetic resonance guided high intensity focused ultrasound and temperature sensitive liposomes in a rabbit Vx2 tumor model. J Control Release 2012; 158:487-94. [PMID: 22210162 PMCID: PMC3319290 DOI: 10.1016/j.jconrel.2011.12.011] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 12/21/2022]
Abstract
Clinical-grade doxorubicin encapsulated low temperature sensitive liposomes (LTSLs) were combined with a clinical magnetic resonance-guided high intensity focused ultrasound (MR-HIFU) platform to investigate in vivo image-guided drug delivery. Plasma pharmacokinetics were determined in 3 rabbits. Fifteen rabbits with Vx2 tumors within superficial thigh muscle were randomly assigned into three treatment groups: 1) free doxorubicin, 2) LTSL and 3) LTSL + MR-HIFU. For the LTSL + MR-HIFU group, mild hyperthermia (40-41 °C) was applied to the tumors using an MR-HIFU system. Image-guided non-invasive hyperthermia was applied for a total of 30 min, completed within 1h after LTSL infusion. High-pressure liquid chromatography (HPLC) analysis of the harvested tumor and organ/tissue homogenates was performed to determine doxorubicin concentration. Fluorescence microscopy was performed to determine doxorubicin spatial distribution in the tumors. Sonication of Vx2 tumors resulted in accurate (mean = 40.5 ± 0.1 °C) and spatially homogenous (SD = 1.0 °C) temperature control in the target region. LTSL + MR-HIFU resulted in significantly higher tumor doxorubicin concentrations (7.6- and 3.4-fold greater compared to free doxorubicin and LTSL respectively, p<0.05, Newman-Keuls). This improved tumor concentration was achieved despite heating <25% of the tumor volume. Free doxorubicin and LTSL treatments appeared to deliver more drug in the tumor periphery as compared to the tumor core. In contrast, LTSL + MR-HIFU treatment suggested an improved distribution with doxorubicin found in both the tumor periphery and core. Doxorubicin bio-distribution in non-tumor organs/tissues was fairly similar between treatment groups. This technique has potential for clinical translation as an image-guided method to deliver drug to a solid tumor.
Collapse
Affiliation(s)
- Ashish Ranjan
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Genevieve Jacobs
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - David L. Woods
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ayele H. Negussie
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ari Partanen
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
- Philips Healthcare, Cleveland, OH
- University of Helsinki, Helsinki, Finland
| | - Pavel S. Yarmolenko
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
- Biomedical Engineering, Duke University, Durham, NC
| | - Carmen E. Gacchina
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Karun V. Sharma
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Victor Frenkel
- Department of Biomedical Engineering, Catholic University of America, Washington, DC
| | - Bradford J. Wood
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Matthew R. Dreher
- Center for Interventional Oncology, Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
68
|
Zhang Y, Peng Z, Chen M, Liu F, Huang J, Xu L, Zhang Y, Chen M. Elevated neutrophil to lymphocyte ratio might predict poor prognosis for colorectal liver metastasis after percutaneous radiofrequency ablation. Int J Hyperthermia 2012; 28:132-40. [DOI: 10.3109/02656736.2011.654374] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
69
|
Wood BJ, Poon RT, Locklin JK, Dreher MR, Ng KK, Eugeni M, Seidel G, Dromi S, Neeman Z, Kolf M, Black CDV, Prabhakar R, Libutti SK. Phase I study of heat-deployed liposomal doxorubicin during radiofrequency ablation for hepatic malignancies. J Vasc Interv Radiol 2012; 23:248-55.e7. [PMID: 22178041 PMCID: PMC3264789 DOI: 10.1016/j.jvir.2011.10.018] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 10/19/2011] [Accepted: 10/21/2011] [Indexed: 01/10/2023] Open
Abstract
PURPOSE A phase I dose escalation study was performed with systemically delivered lyso-thermosensitive liposomal doxorubicin (LTLD). The primary objectives were to determine the safe maximum tolerated dose (MTD), pharmacokinetic properties, and dose-limiting toxicity (DLT) of LTLD during this combination therapy. MATERIALS AND METHODS Subjects eligible for percutaneous or surgical radiofrequency (RF) ablation with primary (n = 9) or metastatic (n = 15) tumors of the liver, with four or fewer lesions as large as 7 cm in diameter, were included. RF ablation was initiated 15 minutes after starting a 30-minute intravenous LTLD infusion. Dose levels between 20 mg/m(2) and 60 mg/m(2) were evaluated. Magnetic resonance imaging, positron emission tomography, and computed tomography were performed at predetermined intervals before and after treatment until evidence of recurrence was seen, administration of additional antitumor treatment was performed, or a total of 3 years had elapsed. RESULTS DLT criteria were met at 60 mg/m(2), and the MTD was defined as 50 mg/m(2). RF ablation was performed during the peak of the plasma concentration-time curve in an effort to yield maximal drug deposition. LTLD produced reversible, dose-dependent neutropenia and leukopenia. CONCLUSIONS LTLD can be safely administered systemically at the MTD (50 mg/m(2)) in combination with RF ablation, with limited and manageable toxicity. Further evaluation of this agent combined with RF ablation is warranted to determine its role in the management of liver tumors.
Collapse
Affiliation(s)
- Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pk., Building 10, Room 1C364, MSC 1182, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Goldberg SN. Science to practice: Which approaches to combination interventional oncologic therapy hold the greatest promise of obtaining maximal clinical benefit? Radiology 2012; 261:667-9. [PMID: 22095989 DOI: 10.1148/radiol.111906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Soundararajan et al (1) were able to demonstrate that combination therapy of radiofrequency (RF) ablation with rhenium 186 (186Re)--labeled liposomal doxorubicin resulted in better drug uptake and reduced tumor growth compared with other therapies tested in a rodent head and neck tumor model. In essence, this “triple” combination therapy of chemotherapy, radiation, and thermal ablation induced significantly smaller viable tumor volume compared with the therapies tested alone at histopathologic examination.
Collapse
Affiliation(s)
- S Nahum Goldberg
- Department of Radiology, Image-guided Therapy and Interventional Oncology Unit, Hadassah Hebrew University Medical Center Jerusalem, Israel.
| |
Collapse
|
71
|
Li C, Krishnan J, Stebbing J, Xu XY. Use of mathematical models to understand anticancer drug delivery and its effect on solid tumors. Pharmacogenomics 2012; 12:1337-48. [PMID: 21919608 DOI: 10.2217/pgs.11.71] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The transport of anticancer drugs and their effect on tumor cells involve a number of physical and biochemical processes. Mathematical modeling provides a tool to help us understand the interaction of these complex processes, thereby contributing to the improvement and optimization of drug delivery. This article starts with a discussion of the biological and physiological properties of tumors, which are often found as barriers to anticancer drug transport and effect. A broad spectrum of mathematical models is reviewed to give an overview of the current state of modeling approaches and different categories of models are outlined. These include pharmacokinetic and transport-based models for the prediction of temporal and temporal-spatial profiles of antidrug concentrations, as well as empirical or deterministic models to describe the effect of drug. We conclude that the systematic elucidation and integration of cellular signal transduction with the biophysical aspects of drug transport will lead to a better understanding of the entire drug-delivery process.
Collapse
Affiliation(s)
- Cong Li
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW72AZ, UK
| | | | | | | |
Collapse
|
72
|
Ahmed M, Moussa M, Goldberg SN. Synergy in cancer treatment between liposomal chemotherapeutics and thermal ablation. Chem Phys Lipids 2011; 165:424-37. [PMID: 22197685 DOI: 10.1016/j.chemphyslip.2011.12.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 01/03/2023]
Abstract
Minimally invasive image-guided tumor ablation using short duration heating via needle-like applicators using energies such as radiofrequency or microwave has seen increasing clinical use to treat focal liver, renal, breast, bone, and lung tumors. Potential benefits of this thermal therapy include reduced morbidity and mortality compared to standard surgical resection and ability to treat non-surgical patients. However, improvements to this technique are required as achieving complete ablation in many cases can be challenging particularly at margins of tumors>3 cm in diameter and adjacent to blood vessels. Thus, one very promising strategy has been to combine thermal tumor ablation with adjuvant nanoparticle-based chemotherapy agents to improve efficiency. Here, we will primarily review principles of thermal ablation to provide a framework for understanding the mechanisms of combination therapy, and review the studies on combination therapy, including presenting preliminary data on the role of such variables as nanoparticle size and thermal dose on improving combination therapy outcome. We will discuss how thermal ablation can also be used to improve overall intratumoral drug accumulation and nanoparticle content release. Finally, in this article we will further describe the appealing off-shoot approach of utilizing thermal ablation techniques not as the primary treatment, but rather, as a means to improve efficiency of intratumoral nanoparticle drug delivery.
Collapse
Affiliation(s)
- Muneeb Ahmed
- Minimally Invasive Tumor Therapy Laboratory, Section of Interventional Radiology, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA.
| | | | | |
Collapse
|
73
|
|
74
|
Negussie AH, Yarmolenko PS, Partanen A, Ranjan A, Jacobs G, Woods D, Bryant H, Thomasson D, Dewhirst MW, Wood BJ, Dreher MR. Formulation and characterisation of magnetic resonance imageable thermally sensitive liposomes for use with magnetic resonance-guided high intensity focused ultrasound. Int J Hyperthermia 2011; 27:140-55. [PMID: 21314334 PMCID: PMC3417228 DOI: 10.3109/02656736.2010.528140] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Objectives of this study were to: 1) develop iLTSL, a low temperature sensitive liposome co-loaded with an MRI contrast agent (ProHance® Gd-HP-DO3A) and doxorubicin, 2) characterise doxorubicin and Gd-HP-DO3A release from iLTSL and 3) investigate the ability of magnetic resonance-guided high intensity focused ultrasound (MR-HIFU) to induce and monitor iLTSL content release in phantoms and in vivo. METHODS iLTSL was passively loaded with Gd-HP-DO3A and actively loaded with doxorubicin. Doxorubicin and Gd-HP-DO3A release was quantified by fluorescence and spectroscopic techniques, respectively. Release with MR-HIFU was examined in tissue-mimicking phantoms containing iLTSL and in a VX2 rabbit tumour model. RESULTS iLTSL demonstrated consistent size and doxorubicin release kinetics after storage at 4°C for 7 days. Release of doxorubicin and Gd-HP-DO3A from iLTSL was minimal at 37°C but fast when heated to 41.3°C. The magnitude of release was not significantly different between doxorubicin and Gd-HP-DO3A over 10 min in HEPES buffer and plasma at 37°, 40° and 41.3°C (p > 0.05). Relaxivity of iLTSL increased significantly (p < 0.0001) from 1.95 ± 0.05 to 4.01 ± 0.1 mMs⁻¹ when heated above the transition temperature. Signal increase corresponded spatially and temporally to MR-HIFU-heated locations in phantoms. Signal increase was also observed in vivo after iLTSL injection and after each 10-min heating (41°C), with greatest increase in the heated tumour region. CONCLUSION An MR imageable liposome formulation co-loaded with doxorubicin and an MR contrast agent was developed. Stability, imageability, and MR-HIFU monitoring and control of content release suggest that MR-HIFU combined with iLTSL may enable real-time monitoring and spatial control of content release.
Collapse
Affiliation(s)
- Ayele H Negussie
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Staruch R, Chopra R, Hynynen K. Localised drug release using MRI-controlled focused ultrasound hyperthermia. Int J Hyperthermia 2010; 27:156-71. [PMID: 21158487 DOI: 10.3109/02656736.2010.518198] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Thermosensitive liposomes provide a mechanism for triggering the local release of anticancer drugs, but this technology requires precise temperature control in targeted regions with minimal heating of surrounding tissue. The objective of this study was to evaluate the feasibility of using MRI-controlled focused ultrasound (FUS) and thermosensitive liposomes to achieve thermally mediated localised drug delivery in vivo. MATERIALS AND METHODS Results are reported from ten rabbits, where a FUS beam was scanned in a circular trajectory to heat 10-15 mm diameter regions in normal thigh to 43°C for 20-30 min. MRI thermometry was used for closed-loop feedback control to achieve temporally and spatially uniform heating. Lyso-thermosensitive liposomal doxorubicin was infused intravenously during hyperthermia. Unabsorbed liposomes were flushed from the vasculature by saline perfusion 2 h later, and tissue samples were harvested from heated and unheated thigh regions. The fluorescence intensity of the homogenised samples was used to calculate the concentration of doxorubicin in tissue. RESULTS Closed-loop control of FUS heating using MRI thermometry achieved temperature distributions with mean, T90 and T10 of 42.9°C, 41.0°C and 44.8°C, respectively, over a period of 20 min. Doxorubicin concentrations were significantly higher in tissues sampled from heated than unheated regions of normal thigh muscle (8.3 versus 0.5 ng/mg, mean per-animal difference = 7.8 ng/mg, P < 0.05, Wilcoxon matched pairs signed rank test). CONCLUSIONS The results show the potential of MRI-controlled focused ultrasound hyperthermia for enhanced local drug delivery with temperature-sensitive drug carriers.
Collapse
Affiliation(s)
- Robert Staruch
- Centre for Research in Image-Guided Therapeutics, Sunnybrook Health Sciences Centre, Toronto, Canada.
| | | | | |
Collapse
|