201
|
Huang CY, Pourgholami MH, Allen BJ. Optimizing radioimmunoconjugate delivery in the treatment of solid tumor. Cancer Treat Rev 2012; 38:854-60. [DOI: 10.1016/j.ctrv.2011.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 02/08/2023]
|
202
|
Ait-Oudhia S, Straubinger RM, Mager DE. Systems pharmacological analysis of paclitaxel-mediated tumor priming that enhances nanocarrier deposition and efficacy. J Pharmacol Exp Ther 2012; 344:103-12. [PMID: 23115220 DOI: 10.1124/jpet.112.199109] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Paclitaxel (PAC)-mediated apoptosis decompresses and primes tumors for enhanced deposition of nanoparticulate agents such as pegylated liposomal doxorubicin (DXR). A quantitative pharmacokinetic/pharmacodynamic (PK/PD) approach was developed to analyze efficacy and identify optima for PAC combined with sterically stabilized liposome (SSL)-DXR. Using data extracted from diverse literature sources, Cremophor-paclitaxel (Taxol(®)) PK was described by a carrier-mediated dispositional model and SSL-DXR PK was described by a two-compartment model with first-order drug release. A hybrid-physiologic, well-stirred model with partition coefficients (Kp) captured intratumor concentrations. Apoptotic responses driving tumor priming were modeled using nonlinear, time-dependent transduction functions. The tumor growth model used net first-order growth and death rate constants, and two transit compartments that captured the temporal displacement of tumor exposure versus effect, and apoptotic signals from each agent were used to drive cytotoxic effects of the combination. The final model captured plasma and intratumor PK data, apoptosis induction profiles, and tumor growth for all treatments/sequences. A feedback loop representing PAC-induced apoptosis effects on Kp(_DXR) enabled the model to capture tumor-priming effects. Simulations to explore time- and sequence-dependent effects of priming indicated that PAC priming increased K(p_DXR) 3-fold. The intratumor concentrations producing maximal and half-maximal effects were 18 and 7.2 μg/ml for PAC, and 17.6 and 14.3 μg/ml for SSL-DXR. The duration of drug-induced apoptosis was 27.4 h for PAC and 15.8 h for SSL-DXR. Simulations suggested that PAC administered 24 h before peak priming could increase efficacy 2.5-fold over experimentally reported results. The quantitative approach developed in this article is applicable for evaluating tumor-priming strategies using diverse agents.
Collapse
Affiliation(s)
- Sihem Ait-Oudhia
- Department of Pharmaceutical Sciences, 456 Kapoor Hall, University at Buffalo, State University of New York, Buffalo, NY 14214, USA.
| | | | | |
Collapse
|
203
|
Perche F, Patel NR, Torchilin VP. Accumulation and toxicity of antibody-targeted doxorubicin-loaded PEG-PE micelles in ovarian cancer cell spheroid model. J Control Release 2012; 164:95-102. [PMID: 22974689 DOI: 10.1016/j.jconrel.2012.09.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 08/28/2012] [Accepted: 09/04/2012] [Indexed: 01/01/2023]
Abstract
We describe the evaluation of doxorubicin-loaded PEG-PE micelles targeting using an ovarian cancer cell spheroid model. Most ovarian cancer patients present at an advanced clinical stage and develop resistance to standard of care platinum/taxane therapy. Doxorubicin is also approved for ovarian cancer but had limited benefits in refractory patients. In this study, we used drug-resistant spheroid cultures of ovarian carcinoma to evaluate the uptake and cytotoxicity of an antibody-targeted doxorubicin formulation. Doxorubicin was encapsulated in polyethylene glycol-phosphatidyl ethanolamine (PEG-PE) conjugated micelles. The doxorubicin-loaded PEG-PE micelles (MDOX) were further decorated with a cancer cell-specific monoclonal 2C5 antibody to obtain doxorubicin-loaded immunomicelles (2C5-MDOX). Targeting and resulting toxicity of doxorubicin-loaded PEG-PE micelles were evaluated in three dimensional cancer cell spheroids. Superior accumulation of 2C5-MDOX compared to free doxorubicin or untargeted MDOX in spheroids was evidenced both by flow cytometry, fluorescence and confocal microscopy. Interestingly, even higher toxicity was measured by lactate dehydrogenase release and terminal deoxynucleotidyl transferase dUTP nick end labeling of targeted doxorubicin micelles in Bcl-2 overexpressing adriamycin-resistant spheroids. Overall, these results support use of spheroids to evaluate tumor targeted drug delivery.
Collapse
Affiliation(s)
- Federico Perche
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | | | | |
Collapse
|
204
|
Quantitative immunohistochemistry for evaluating the distribution of Ki67 and other biomarkers in tumor sections and use of the method to study repopulation in xenografts after treatment with paclitaxel. Neoplasia 2012; 14:324-34. [PMID: 22577347 DOI: 10.1593/neo.12346] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 03/20/2012] [Accepted: 03/21/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Surviving cells may repopulate tumors between courses of chemotherapy, thereby reducing the effectiveness of treatment. Using a novel quantitative method, we characterize the influence of the tumor microenvironment on repopulation of surviving cells in human tumor xenografts after paclitaxel treatment and evaluate the potential of gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, to inhibit repopulation. METHODS High-EGFR-expressing A431 xenografts and low-EGFR-expressing MCF-7 xenografts were treated with paclitaxel or gefitinib. Time-dependent changes in cell proliferation (Ki67) and apoptosis (cleaved caspase 3) in relation to total and functional tumor blood vessels (recognized by CD31 and a flow marker), and regions of hypoxia (recognized by EF5) were quantified using fluorescence microscopy. RESULTS Decrease in functional tumor vasculature and in cell proliferation and increase in apoptosis were observed in A431 xenografts after treatment with either paclitaxel or gefitinib. There was a rebound in functional vasculature and cell proliferation ≈ 12 days after treatment with paclitaxel, and repopulation was observed from tumor cells close to regions of hypoxia. Cell proliferation increased ≈ 5 days after the last dose of gefitinib. There were minimal effects of paclitaxel or gefitinib on cell proliferation, cell death, or tumor vasculature in MCF-7 xenografts. CONCLUSIONS Repopulation in A431 xenografts after treatment with paclitaxel was associated with changes in functional tumor vasculature. Gefitinib decreased cell proliferation in EGFR-overexpressing tumor xenografts, suggesting its potential to inhibit repopulation when used in sequence with chemotherapy.
Collapse
|
205
|
Guo P, Haque F, Hallahan B, Reif R, Li H. Uniqueness, advantages, challenges, solutions, and perspectives in therapeutics applying RNA nanotechnology. Nucleic Acid Ther 2012. [PMID: 22913595 DOI: 10.1201/b15152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
The field of RNA nanotechnology is rapidly emerging. RNA can be manipulated with the simplicity characteristic of DNA to produce nanoparticles with a diversity of quaternary structures by self-assembly. Additionally RNA is tremendously versatile in its function and some RNA molecules display catalytic activities much like proteins. Thus, RNA has the advantage of both worlds. However, the instability of RNA has made many scientists flinch away from RNA nanotechnology. Other concerns that have deterred the progress of RNA therapeutics include the induction of interferons, stimulation of cytokines, and activation of other immune systems, as well as short pharmacokinetic profiles in vivo. This review will provide some solutions and perspectives on the chemical and thermodynamic stability, in vivo half-life and biodistribution, yield and production cost, in vivo toxicity and side effect, specific delivery and targeting, as well as endosomal trapping and escape.
Collapse
Affiliation(s)
- Peixuan Guo
- Nanobiotechnology Center, Markey Cancer Center and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | | | | | | | |
Collapse
|
206
|
Creixell M, Peppas NA. Co-delivery of siRNA and therapeutic agents using nanocarriers to overcome cancer resistance. NANO TODAY 2012; 7:367-379. [PMID: 26257819 PMCID: PMC4527553 DOI: 10.1016/j.nantod.2012.06.013] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
There are two main mechanisms by which cells become multidrug resistant (MDR): by increasing drug efflux pumps on the cell membrane and by increasing anti-apoptotic pathways. The use of nanotechnology to develop nanodelivery systems has allowed researchers to overcome limitations of antineoplastic drugs by increasing the solubility of the drug and decreasing the toxicity to healthy tissues. By encapsulating drugs into nanoparticles that bypass the efflux pumps, drug efflux is reduced, hence increasing the intracellular concentration of the drug. siRNA has the ability to disrupt cellular pathways by knocking down genes, opening the door to down regulating anti-apoptotic pathways. The use of nanocarriers to deliver siRNA, prevents both renal clearance and RNase degradation by protecting siRNA chains, increasing their half life in blood. It has been suggested that co-delivering drugs and siRNA together in the same delivery system would be more effective in overcoming resistance of cancer cells than co-treatment of cancer cells with delivery systems carrying either siRNA or drugs. In this study we discuss the progress of nanoscale co-delivery systems in overcoming multidrug cancer resistance.
Collapse
Affiliation(s)
- Mar Creixell
- Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 78712, USA
| | - Nicholas A. Peppas
- Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, C0800, The University of Texas at Austin, Austin, TX 78712, USA
- College of Pharmacy, C0400, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
207
|
Torosean S, Flynn B, Axelsson J, Gunn J, Samkoe KS, Hasan T, Doyley MM, Pogue BW. Nanoparticle uptake in tumors is mediated by the interplay of vascular and collagen density with interstitial pressure. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 9:151-8. [PMID: 22841910 DOI: 10.1016/j.nano.2012.07.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/24/2012] [Accepted: 07/12/2012] [Indexed: 11/30/2022]
Abstract
UNLABELLED Nanoparticle delivery into solid tumors is affected by vessel density, interstitial fluid pressure (IFP) and collagen, as shown in this article by contrasting the in vivo macroscopic quantitative uptake of 40 nm fluorescent beads in three tumor types.The fluorescence uptake was quantified on individual animals by normalization with the transmitted light and then normalized to normal tissue uptake in each mouse. Mean data for uptake in individual tumor lines then showed expected trends with the largest uptake in the most vascularized tumor line. Tumor lines with increased collagen were also consistent with highest interstitial fluid pressure and correlated with lowest uptake of nanoparticles. The data is consistent with a delivery model indicating that while vascular permeability is maximized by neovascular growth, it is inhibited by collagen content and the resulting interstitial pressure. Imaging of these parameters in vivo can lead to better individual noninvasive methods to assess drug penetration in situ. FROM THE CLINICAL EDITOR In this manuscript the dependence of nanoparticle delivery is addressed from the standpoint of vascular factors (the more vascularized, the better delivery) and as a function of collagen density and interstitial pressure (the higher these are, the worse the delivery).
Collapse
Affiliation(s)
- Sason Torosean
- Department of Physics and Astronomy, Dartmouth College, Hanover, New Hampshire, USA
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Madaan A, Singh P, Awasthi A, Verma R, Singh AT, Jaggi M, Mishra SK, Kulkarni S, Kulkarni H. Efficiency and mechanism of intracellular paclitaxel delivery by novel nanopolymer-based tumor-targeted delivery system, NanoxelTM. Clin Transl Oncol 2012; 15:26-32. [DOI: 10.1007/s12094-012-0883-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/27/2012] [Indexed: 12/15/2022]
|
209
|
Roy Chaudhuri T, Arnold RD, Yang J, Turowski SG, Qu Y, Spernyak JA, Mazurchuk R, Mager DE, Straubinger RM. Mechanisms of tumor vascular priming by a nanoparticulate doxorubicin formulation. Pharm Res 2012; 29:3312-24. [PMID: 22798260 DOI: 10.1007/s11095-012-0823-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/25/2012] [Indexed: 11/26/2022]
Abstract
PURPOSE Tumor vascular normalization by antiangiogenic agents may increase tumor perfusion but reestablish vascular barrier properties in CNS tumors. Vascular priming via nanoparticulate carriers represents a mechanistically distinct alternative. This study investigated mechanisms by which sterically-stabilized liposomal doxorubicin (SSL-DXR) modulates tumor vascular properties. METHODS Functional vascular responses to SSL-DXR were investigated in orthotopic rat brain tumors using deposition of fluorescent permeability probes and dynamic contrast-enhanced magnetic resonance imaging. Microvessel density and tumor burden were quantified by immunohistochemistry (CD-31) and quantitative RT-PCR (VE-cadherin). RESULTS Administration of SSL-DXR (5.7 mg/kg iv) initially (3-4 days post-treatment) decreased tumor vascular permeability, k(trans) (vascular exchange constant), vascular endothelial cell content, microvessel density, and deposition of nanoparticulates. Tumor vasculature became less chaotic. Permeability and perfusion returned to control values 6-7 days post-treatment, but intratumor SSL-DXR depot continued to effect tumor vascular endothelial compartment 7-10 days post-treatment, mediating enhanced permeability. CONCLUSIONS SSL-DXR ultimately increased tumor vascular permeability, but initially normalized tumor vasculature and decreased tumor perfusion, permeability, and nanoparticulate deposition. These temporal changes in vascular integrity resulting from a single SSL-DXR dose have important implications for the design of combination therapies incorporating nanoparticle-based agents for tumor vascular priming.
Collapse
Affiliation(s)
- Tista Roy Chaudhuri
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 445 Kapoor Hall, Buffalo, New York 14214-8033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Bazylińska U, Pietkiewicz J, Saczko J, Nattich-Rak M, Rossowska J, Garbiec A, Wilk KA. Nanoemulsion-templated multilayer nanocapsules for cyanine-type photosensitizer delivery to human breast carcinoma cells. Eur J Pharm Sci 2012; 47:406-20. [PMID: 22796218 DOI: 10.1016/j.ejps.2012.06.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/25/2012] [Accepted: 06/25/2012] [Indexed: 10/28/2022]
Abstract
There is great clinical interest in developing novel nanocarriers for hydrophobic cyanine dyes used as photosensitizing agents in photodynamic therapy (PDT). In the present study we have employed nanoemulsion-templated oil-core multilayer nanocapsules as robust nanocarriers for a cyanine-type photosensitizer IR-786. These nanoproducts were fabricated via layer-by-layer (LbL) adsorption of oppositely charged polyelectrolytes (PEs), i.e., anionic PSS and cationic PDADMAC on nanoemulsion liquid cores created by dicephalic or bulky saccharide-derived cationic surfactants. All nanocapsules, with different thicknesses of the PE shell and average size <200 nm (measured by DLS) demonstrated good capacity for IR-786 encapsulation. The nanocarriers were visualized by SEM and AFM and their photo-induced anticancer effect and cellular internalization in human breast carcinoma MCF-7/WT cells were determined. Biological response of the cell culture, expressed as dark and photocytotoxicity as well as fluorescence of drug molecules loaded in the multilayer vehicles, analyzed by the FACS and CLSM techniques, have indicated that the delivered IR-786 did not aggregate inside the cells and could, therefore, act as an effective third-generation photosensitizing agent. In vitro biological experiments demonstrated that the properties of studied nanostructures depended upon the PE type and the envelope thickness as well as on the surfactant architecture in the nanoemulsion-based templates employed for the nanocapsule fabrication. Similarity of results obtained for stored (three weeks in the dark at room temperature) and freshly-prepared nanocapsules, attests to viability of this stable, promising drug delivery system for poorly water-soluble cyanines useful in PDT.
Collapse
Affiliation(s)
- Urszula Bazylińska
- Faculty of Chemistry, Wroclaw University of Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | | | | | | | | | | | | |
Collapse
|
211
|
Spugnini EP, Fanciulli M, Citro G, Baldi A. Preclinical models in electrochemotherapy: the role of veterinary patients. Future Oncol 2012; 8:829-37. [DOI: 10.2217/fon.12.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Electrochemotherapy is a tumor treatment that adapts the systemic or local delivery of anticancer drugs by the application of permeabilizing electric pulses with appropriate amplitude and waveforms. This allows the use of lipophobic drugs, which frequently have a narrow therapeutic index, with a decreased morbidity for the patient, while maintaining appropriate anticancer efficacy. Electrochemotherapy is used in humans for the treatment of cutaneous neoplasms or the palliation of skin tumor metastases, and a standard operating procedure has been devised. In veterinary oncology, the electrochemotherapy approach is gaining popularity, becoming a first-line treatment in consideration of its high efficacy and low toxicity. This review summarizes the state of the art in veterinary oncology as a preclinical model.
Collapse
Affiliation(s)
| | - Maurizio Fanciulli
- SAFU Department, Regina Elena Cancer Institute, Via delle Messi d’Oro 156, Rome 00158, Italy
| | | | | |
Collapse
|
212
|
Pak Y, Zhang Y, Pastan I, Lee B. Antigen shedding may improve efficiencies for delivery of antibody-based anticancer agents in solid tumors. Cancer Res 2012; 72:3143-52. [PMID: 22562466 PMCID: PMC3408876 DOI: 10.1158/0008-5472.can-11-3925] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recombinant immunotoxins (RIT) are targeted anticancer agents that are composed of a targeting antibody fragment and a protein toxin fragment. SS1P is a RIT that targets mesothelin on the surface of cancer cells and is being evaluated in patients with mesothelioma. Mesothelin, like many other target antigens, is shed from the cell surface. However, whether antigen shedding positively or negatively affects the delivery of RIT remains unknown. In this study, we used experimental data with SS1P to develop a mathematical model that describes the relationship between tumor volume changes and the dose level of the administered RIT, while accounting for the potential effects of antigen shedding.
Collapse
Affiliation(s)
- Youngshang Pak
- Department of Chemistry and Institute of Functional Materials, Pusan National University, Busan 609-735, Republic of Korea
| | - Yujian Zhang
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | - Ira Pastan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | - Byungkook Lee
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| |
Collapse
|
213
|
Zhou F, Hu J, Shao JH, Zou SB, Shen SL, Luo ZQ. Metronomic chemotherapy in combination with antiangiogenic treatment induces mosaic vascular reduction and tumor growth inhibition in hepatocellular carcinoma xenografts. J Cancer Res Clin Oncol 2012; 138:1879-90. [PMID: 22736027 DOI: 10.1007/s00432-012-1270-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 06/13/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND In addition to sprouting angiogenesis, other mechanisms, such as mosaic tumor vessel formation, have been recognized to contribute to tumor vascularization. We sought to examine vascular alteration as well as tumor growth inhibition after treatment with antiangiogenic therapy, chemotherapy alone or in combination. METHODS Hepatocellular carcinoma cells (Hep3B) expressed green fluorescent protein were utilized to establish orthotopic xenograft model in nude mice. The formation and distribution of mosaic vessels was analyzed quantitatively by immunolabeling. Next, changes in tumor microcirculation and therapeutic effects on tumor growth were evaluated in several different treatment groups: control, conventional doxorubicin, metronomic doxorubicin, bevacizumab, bevacizumab plus conventional doxorubicin, and bevacizumab plus metronomic doxorubicin. In addition, we examined the effects of combined regimens on lung metastasis using a highly metastatic human hepatocellular carcinoma (HCCLM3) mouse model. RESULTS Approximately 62 % of the vessels were present in the central part or near the midsection of the tumor and were mosaic. Only the combined antiangiogenic treatment and chemotherapy (metronomic schedule, P = 0.00; conventional schedule, P = 0.02) had a significant effect on the degree of mosaic vasculature. Metronomic doxorubicin in combination with bevacizumab had an even more profound effect than bevacizumab plus conventional doxorubicin (P < 0.05) on tumor growth inhibition and survival. However, bevacizumab plus metronomic doxorubicin failed to inhibit lung metastasis compared with antiangiogenic monotherapy. CONCLUSIONS Metronomic chemotherapy in combination with antiangiogenic treatment results in the reduction of mosaic tumor vasculature, inhibition of tumor growth, and enhanced survival of mice. Further investigation of drug scheduling is required to optimize antitumor activity.
Collapse
MESH Headings
- Administration, Metronomic
- Angiogenesis Inhibitors/administration & dosage
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bevacizumab
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Doxorubicin/administration & dosage
- Drug Administration Schedule
- Humans
- Injections, Intravenous
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Survival Analysis
- Treatment Outcome
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Fan Zhou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 1 Mingde Road, Nanchang 330006, China.
| | | | | | | | | | | |
Collapse
|
214
|
Glebova KV, Marakhonov AV, Baranova AV, Skoblov MY. Therapeutic siRNAs and nonviral systems for their delivery. Mol Biol 2012. [DOI: 10.1134/s0026893312020069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
215
|
Wang J, Lu Z, Gao Y, Wientjes MG, Au JLS. Improving delivery and efficacy of nanomedicines in solid tumors: role of tumor priming. Nanomedicine (Lond) 2012; 6:1605-20. [PMID: 22077464 DOI: 10.2217/nnm.11.141] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Effectiveness of nanomedicines in cancer therapy is limited in part by inadequate delivery and transport in tumor interstitium. This article reviews the experimental approaches to improve nanomedicine delivery and transport in solid tumors. These approaches include tumor vasculature normalization, interstitial fluid pressure modulation, enzymatic extracellular matrix degradation, and apoptosis-inducing tumor priming technology. We advocate the latter approach due to its ease and practicality (accomplished with standard-of-care chemotherapy, such as paclitaxel) and tumor selectivity. Examples of applying tumor priming to deliver nanomedicines and to design drug/RNAi-loaded carriers are discussed.
Collapse
Affiliation(s)
- Jie Wang
- Optimum Therapeutics LLC, The Ohio State University Science Tech Village, Columbus, OH 43212, USA
| | | | | | | | | |
Collapse
|
216
|
Lin X, Gao R, Zhang Y, Qi N, Zhang Y, Zhang K, He H, Tang X. Lipid nanoparticles for chemotherapeutic applications: strategies to improve anticancer efficacy. Expert Opin Drug Deliv 2012; 9:767-81. [DOI: 10.1517/17425247.2012.685933] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
217
|
Wong H, Vernillet L, Peterson A, Ware JA, Lee L, Martini JF, Yu P, Li C, Del Rosario G, Choo EF, Hoeflich KP, Shi Y, Aftab BT, Aoyama R, Lam ST, Belvin M, Prescott J. Bridging the gap between preclinical and clinical studies using pharmacokinetic-pharmacodynamic modeling: an analysis of GDC-0973, a MEK inhibitor. Clin Cancer Res 2012; 18:3090-9. [PMID: 22496205 DOI: 10.1158/1078-0432.ccr-12-0445] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE GDC-0973 is a potent and selective mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor. Pharmacokinetic-pharmacodynamic (PK-PD) modeling was used to relate GDC-0973 plasma and tumor concentrations, tumor pharmacodynamics and antitumor efficacy to establish pharmacokinetic endpoints and predict active doses in the clinic. EXPERIMENTAL DESIGN A PK-PD model was used to characterize GDC-0973 tumor disposition and in vivo potency in WM-266-4 xenograft mice. Simulations were conducted using the PK-PD model along with human pharmacokinetics to identify a target plasma concentration and predict active doses. In vivo potency and antitumor efficacy were characterized in A375 melanoma xenograft mice, and a population-based integrated PK-PD-efficacy model was used to relate tumor pharmacodynamics (%pERK decrease) to antitumor activity. RESULTS GDC-0973 showed a sustained tumor pharmacodynamic response due to longer residence in tumor than in plasma. Following single doses of GDC-0973, estimated in vivo IC(50) values of %pERK decrease based on tumor concentrations in xenograft mice were 0.78 (WM-266-4) and 0.52 μmol/L (A375). Following multiple doses of GDC-0973, the estimated in vivo IC(50) value in WM-266-4 increased (3.89 μmol/L). Human simulations predicted a minimum target plasma concentration of 83 nmol/L and an active dose range of 28 to 112 mg. The steep relationship between tumor pharmacodynamics (%pERK decrease) and antitumor efficacy suggests a pathway modulation threshold beyond which antitumor efficacy switches on. CONCLUSIONS Clinical observations of %pERK decrease and antitumor activity were consistent with model predictions. This article illustrates how PK-PD modeling can improve the translation of preclinical data to humans by providing a means to integrate preclinical and early clinical data.
Collapse
Affiliation(s)
- Harvey Wong
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, MS 412a, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Kim SY, Kim TH, Choi JH, Lee KC, Park KD, Lee SJ, Kuh HJ. Evaluation of interstitial protein delivery in multicellular layers model. Arch Pharm Res 2012; 35:531-41. [PMID: 22477201 DOI: 10.1007/s12272-012-0317-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 12/24/2022]
Abstract
The limited efficacy of anticancer protein drugs is related to their poor distribution in tumor tissue. We examined interstitial delivery of four model proteins of different molecular size and bioaffinity in multicellular layers (MCL) of human cancer cells. Model proteins were tumor necrosis factor-related apoptosis-including ligand (TRAIL), cetuximab, RNase A, and IgG. MCLs were cultured in Transwell inserts, exposed to drugs, then cryo-sectioned for image acquisition using fluorescence microscopy (fluorescent dye-labeled TRAIL, RNase A, IgG) or immunohistochemistry (cetuximab). TRAIL and cetuximab showed partial penetration into MCLs, whereas RNase A and IgG showed insignificant penetration. At 10-fold higher dose, a significant increase in penetration was observed for IgG only, while cetuximab showed an intense accumulation limited to the front layers. PEGylated TRAIL and RNase A formulated in a heparin-Pluronic (HP) nanogel showed significantly improved penetration attributable to increased stability and extracellular matrix binding, respectively. IgG penetration was significantly enhanced with paclitaxel pretreatment as a penetration enhancer. The present study suggests that MCL culture may be useful in evaluation of protein delivery in the tumor interstitium. Four model proteins showed limited interstitial penetration in MCL cultures. Bioaffinity, rather than molecular size, seems to have a positive effect on tissue penetration, although high binding affinity may lead to sequestration in the front cell layers. Polymer conjugation and nanoformulation, such as PEGylation and HP nanogel, or use of penetration enhancers are potential strategies to increase interstitial delivery of anticancer protein drugs.
Collapse
Affiliation(s)
- Soo-Yeon Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | |
Collapse
|
219
|
Abstract
Anticancer agents continue to be a preferred therapeutic option for several malignancies. Despite their effectiveness, oncologists are continually looking for tumor-specific anticancer agents to prevent adverse effects in patients. Targeting of imaging agents to cancerous tissue is another area that is enthusiastically explored to circumvent some of the drawbacks that current imaging agents possess, including the inability to target small tumor cells, inadequate imaging period, and the risk of renal damage. Formulation scientists have explored nanotechnology-based delivery systems for targeting anticancer agents and tumor-imaging agents to cancer tissue. Targeting with nanotechnology-based delivery systems has been investigated by both passive and active mechanisms with significant clinical success. This review presents a discussion on targeting strategies used for the delivery of nanoparticles by passive and active mechanisms, focusing more specifically on active targeting of nanoparticles using albumin, folic acid, transferrin, and aptamers as targeting ligands.
Collapse
|
220
|
Pink DBS, Schulte W, Parseghian MH, Zijlstra A, Lewis JD. Real-time visualization and quantitation of vascular permeability in vivo: implications for drug delivery. PLoS One 2012; 7:e33760. [PMID: 22479438 PMCID: PMC3315578 DOI: 10.1371/journal.pone.0033760] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/16/2012] [Indexed: 01/09/2023] Open
Abstract
The leaky, heterogeneous vasculature of human tumors prevents the even distribution of systemic drugs within cancer tissues. However, techniques for studying vascular delivery systems in vivo often require complex mammalian models and time-consuming, surgical protocols. The developing chicken embryo is a well-established model for human cancer that is easily accessible for tumor imaging. To assess this model for the in vivo analysis of tumor permeability, human tumors were grown on the chorioallantoic membrane (CAM), a thin vascular membrane which overlays the growing chick embryo. The real-time movement of small fluorescent dextrans through the tumor vasculature and surrounding tissues were used to measure vascular leak within tumor xenografts. Dextran extravasation within tumor sites was selectively enhanced an interleukin-2 (IL-2) peptide fragment or vascular endothelial growth factor (VEGF). VEGF treatment increased vascular leak in the tumor core relative to surrounding normal tissue and increased doxorubicin uptake in human tumor xenografts. This new system easily visualizes vascular permeability changes in vivo and suggests that vascular permeability may be manipulated to improve chemotherapeutic targeting to tumors.
Collapse
Affiliation(s)
| | | | | | - Andries Zijlstra
- Innovascreen, Inc., Halifax, Nova Scotia, Canada
- Department of Pathology, Vanderbilt University, Nashville, Tennesee, United States of America
| | - John D. Lewis
- Innovascreen, Inc., Halifax, Nova Scotia, Canada
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
221
|
Zahedi P, Yoganathan R, Piquette-Miller M, Allen C. Recent advances in drug delivery strategies for treatment of ovarian cancer. Expert Opin Drug Deliv 2012; 9:567-83. [DOI: 10.1517/17425247.2012.665366] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
222
|
Zhao P, Grillaud M, Salmon L, Ruiz J, Astruc D. Click Functionalization of Gold Nanoparticles Using the Very Efficient Catalyst Copper(I) (Hexabenzyl)tris(2-aminoethyl)- amine Bromide. Adv Synth Catal 2012. [DOI: 10.1002/adsc.201100865] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
223
|
Waite CL, Roth CM. Nanoscale drug delivery systems for enhanced drug penetration into solid tumors: current progress and opportunities. Crit Rev Biomed Eng 2012; 40:21-41. [PMID: 22428797 DOI: 10.1615/critrevbiomedeng.v40.i1.20] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Poor penetration of anticancer drags into solid tumors significantly limits their efficacy. This phenomenon has long been observed for small-molecule chemotherapeutics, and it can be even more pronounced for nanoscale therapies. Nanoparticles have enormous potential for the treatment of cancer due to their wide applicability as drug delivery and imaging vehicles and their size-dependent accumulation into solid tumors by the enhanced permeability and retention (EPR) effect. Further, synthetic nanoparticles can be engineered to overcome barriers to drag delivery. Despite their promise for the treatment of cancer, relatively little work has been done to study and improve their ability to diffuse into solid tumors following passive accumulation in the tumor vasculature. In this review, we present the complex issues governing efficient penetration of nanoscale therapies into solid tumors. The current methods available to researchers to study nanoparticle penetration into malignant tumors are described, and the most recent works studying the penetration of nanoscale materials into solid tumors are summarized. We conclude with an overview of the important nanoparticle design parameters governing their tumor penetration, as well as by highlighting critical directions in this field.
Collapse
Affiliation(s)
- Carolyn L Waite
- Department of Chemical and Biochemical Engineering, Rutgers University, New Brunswick, New Jersey, USA
| | | |
Collapse
|
224
|
Erbertseder K, Reichold J, Flemisch B, Jenny P, Helmig R. A coupled discrete/continuum model for describing cancer-therapeutic transport in the lung. PLoS One 2012; 7:e31966. [PMID: 22438873 PMCID: PMC3305605 DOI: 10.1371/journal.pone.0031966] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/16/2012] [Indexed: 11/29/2022] Open
Abstract
We propose a computational simulation framework for describing cancer-therapeutic transport in the lung. A discrete vascular graph model (VGM) is coupled to a double-continuum model (DCM) to determine the amount of administered therapeutic agent that will reach the cancer cells. An alveolar cell carcinoma is considered. The processes in the bigger blood vessels (arteries, arterioles, venules and veins) are described by the VGM. The processes in the alveolar capillaries and the surrounding tissue are represented by a continuum approach for porous media. The system of equations of the coupled discrete/continuum model contains terms that account for degradation processes of the therapeutic agent, the reduction of the number of drug molecules by the lymphatic system and the interaction of the drug with the tissue cells. The functionality of the coupled discrete/continuum model is demonstrated in example simulations using simplified pulmonary vascular networks, which are designed to show-off the capabilities of the model rather than being physiologically accurate.
Collapse
Affiliation(s)
- Karin Erbertseder
- Department of Hydromechanics and Modeling of Hydrosystems, Institute for Modelling Hydraulic and Environmental Systems, University of Stuttgart, Stuttgart, Germany.
| | | | | | | | | |
Collapse
|
225
|
Desai N. Challenges in development of nanoparticle-based therapeutics. AAPS JOURNAL 2012; 14:282-95. [PMID: 22407288 PMCID: PMC3326161 DOI: 10.1208/s12248-012-9339-4] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/17/2012] [Indexed: 02/08/2023]
Abstract
In recent years, nanotechnology has been increasingly applied to the area of drug development. Nanoparticle-based therapeutics can confer the ability to overcome biological barriers, effectively deliver hydrophobic drugs and biologics, and preferentially target sites of disease. However, despite these potential advantages, only a relatively small number of nanoparticle-based medicines have been approved for clinical use, with numerous challenges and hurdles at different stages of development. The complexity of nanoparticles as multi-component three dimensional constructs requires careful design and engineering, detailed orthogonal analysis methods, and reproducible scale-up and manufacturing process to achieve a consistent product with the intended physicochemical characteristics, biological behaviors, and pharmacological profiles. The safety and efficacy of nanomedicines can be influenced by minor variations in multiple parameters and need to be carefully examined in preclinical and clinical studies, particularly in context of the biodistribution, targeting to intended sites, and potential immune toxicities. Overall, nanomedicines may present additional development and regulatory considerations compared with conventional medicines, and while there is generally a lack of regulatory standards in the examination of nanoparticle-based medicines as a unique category of therapeutic agents, efforts are being made in this direction. This review summarizes challenges likely to be encountered during the development and approval of nanoparticle-based therapeutics, and discusses potential strategies for drug developers and regulatory agencies to accelerate the growth of this important field.
Collapse
Affiliation(s)
- Neil Desai
- Strategic Platforms, Abraxis BioScience, 11755 Wilshire Blvd., Suite 2300, Los Angeles, California 90025, USA.
| |
Collapse
|
226
|
A NIR heptamethine dye with intrinsic cancer targeting, imaging and photosensitizing properties. Biomaterials 2012; 33:2230-9. [DOI: 10.1016/j.biomaterials.2011.11.081] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 11/26/2011] [Indexed: 11/20/2022]
|
227
|
Choo EF, Belvin M, Boggs J, Deng Y, Hoeflich KP, Ly J, Merchant M, Orr C, Plise E, Robarge K, Martini JF, Kassees R, Aoyama RG, Ramaiya A, Johnston SH. Preclinical disposition of GDC-0973 and prospective and retrospective analysis of human dose and efficacy predictions. Drug Metab Dispos 2012; 40:919-27. [PMID: 22315332 DOI: 10.1124/dmd.111.043778] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
[3,4-Difluoro-2-(2-fluoro-4-iodo-phenylamino)-phenyl]-((S)-3-hydroxy-3-piperidin-2-yl-azetidin-1-yl)-methanone (GDC-0973) is a potent and highly selective inhibitor of mitogen-activated protein kinase(MAPK)/extracellular signal-regulated kinase (ERK) 1/2 (MEK1/2), a MAPK kinase that activates ERK1/2. The objectives of these studies were to characterize the disposition of GDC-0973 in preclinical species and to determine the relationship of GDC-0973 plasma concentrations to efficacy in Colo205 mouse xenograft models. The clearance (CL) of GDC-0973 was moderate in mouse (33.5 ml · min(-1) · kg(-1)), rat (37.9 ± 7.2 ml · min(-1) · kg(-1)), and monkey (29.6 ± 8.5 ml · min(-1) · kg(-1)). CL in dog was low (5.5 ± 0.3 ml · min(-1) · kg(-1)). The volume of distribution across species was large, 6-fold to 15-fold body water; half-lives ranged from 4 to 13 h. Protein binding in mouse, rat, dog, monkey, and human was high, with percentage unbound, 1 to 6%. GDC-0973-related radioactivity was rapidly and extensively distributed to tissues; however, low concentrations were observed in the brain. In rats and dogs, [(14)C]GDC-0973 was well absorbed (fraction absorbed, 70-80%). The majority of [(14)C]GDC-0973-related radioactivity was recovered in the bile of rat (74-81%) and dog (65%). The CL and volume of distribution of GDC-0973 in human, predicted by allometry, was 2.9 ml · min(-1) · kg(-1) and 9.9 l/kg, respectively. The predicted half-life was 39 h. To characterize the relationship between plasma concentration of GDC-0973 and tumor growth inhibition, pharmacokinetic-pharmacodynamic modeling was applied using an indirect response model. The KC(50) value for tumor growth inhibition in Colo205 xenografts was estimated to be 0.389 μM, and the predicted clinical efficacious dose was ∼10 mg. Taken together, these data are useful in assessing the disposition of GDC-0973, and where available, comparisons with human data were made.
Collapse
Affiliation(s)
- Edna F Choo
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Delivery and biodistribution of siRNA for cancer therapy: challenges and future prospects. Ther Deliv 2012; 3:245-61. [DOI: 10.4155/tde.11.155] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
RNAi-based approaches provide a promising therapeutic modality for the treatment of cancer. The inaccessibility of tumors in different cancer types necessitates the development of safe, specific and efficient systemic delivery systems to meet therapeutic need. The translation of siRNA-based cancer therapeutics to the clinic is hindered by several challenges associated with the cargo (siRNA) and the delivery system, including susceptibility to nucleases; insufficient circulation half-life due to phagocytosis by the reticuloendothelial system, transient and poor biodistribution in the tumor tissue; cellular uptake; inability to escape endosomes and release into the cytosolic compartment for an RNAi-mediated effect; microRNA-like unintended off-target effects; undesirable immune stimulation; and carrier-related toxicity. This review provides an overview of the pharmacokinetic and biodistribution challenges witnessed in the delivery of siRNA when administered systemically. It also describes the current delivery approaches using liposome-, polymer- and peptide-based delivery systems shown to elicit significant gene silencing and tumor growth regression in proof-of-concept studies. As part of future perspectives, delivery agents that showed significant efficacy in preclinical rodent models and clinical trials are also reviewed.
Collapse
|
229
|
Moore LK, Gatica M, Chow EK, Ho D. Diamond-Based Nanomedicine: Enhanced Drug Delivery and Imaging. ACTA ACUST UNITED AC 2012. [DOI: 10.1089/dst.2012.0007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
230
|
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: 1.1] [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
|
231
|
Li Y, Wang J, Wientjes MG, Au JLS. Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor. Adv Drug Deliv Rev 2012; 64:29-39. [PMID: 21569804 DOI: 10.1016/j.addr.2011.04.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 04/17/2011] [Accepted: 04/22/2011] [Indexed: 10/18/2022]
Abstract
Advances in molecular medicines have led to identification of promising targets on cellular and molecular levels. These targets are located in extracellular and intracellular compartments. The latter include cytosol, nucleus, mitochondrion, Golgi apparatus and endoplasmic reticulum. This report gives an overview on the barriers to delivering nanomedicines to various target sites within a solid tumor, the experimental approaches to overcome such barriers, and the potential utility of nanotechnology.
Collapse
|
232
|
Le Duc G, Miladi I, Alric C, Mowat P, Bräuer-Krisch E, Bouchet A, Khalil E, Billotey C, Janier M, Lux F, Epicier T, Perriat P, Roux S, Tillement O. Toward an image-guided microbeam radiation therapy using gadolinium-based nanoparticles. ACS NANO 2011; 5:9566-74. [PMID: 22040385 DOI: 10.1021/nn202797h] [Citation(s) in RCA: 162] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Ultrasmall gadolinium-based nanoparticles (GBNs) induce both a positive contrast for magnetic resonance imaging and a radiosentizing effect. The exploitation of these characteristics leads to a greater increase in lifespan of rats bearing brain tumors since the radiosensitizing effect of GBNs can be activated by X-ray microbeams when the gadolinium content is, at the same time, sufficiently high in the tumor and low in the surrounding healthy tissue. GBNs exhibit therefore an interesting potential for image-guided radiotherapy.
Collapse
Affiliation(s)
- Géraldine Le Duc
- ID17 Biomedical Beamline, European Synchrotron Radiation Facility, 6 Rue Jules Horowitz, 38000 Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Kim JI, Lee BS, Chun C, Cho JK, Kim SY, Song SC. Long-term theranostic hydrogel system for solid tumors. Biomaterials 2011; 33:2251-9. [PMID: 22189146 DOI: 10.1016/j.biomaterials.2011.11.083] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 11/26/2011] [Indexed: 11/26/2022]
Abstract
The long-term theranostic hydrogel system for solid tumors was prepared via simple physical mixing, which consisted of three major parts: the thermosensitive/biodegradable poly(organophosphazene) hydrogel, PEGylated cobalt ferrite nanoparticles, and paclitaxel (PTX). The PEGylated cobalt ferrite nanoparticles showed extremely low cytotoxicity due to the surface modification using PEG chains. The long-term theranostic hydrogel system showed adequate properties to be used for long-term MR theragnosis. In particular, the theranostic hydrogel gradually degraded over 28 days, and the PTX was sustainedly released out from the theranostic hydrogel over the same period in vitro. Furthermore, the in vivo efficacy of long-term MR theragnosis using the theranostic hydrogel system was estimated successfully over 3 weeks by using high field (4.7 T) animal MRI and solid tumor-bearing mice. Based on our results, we expect that this system can supply multiple data regarding a) the progress of therapy and b) the treatment processes via one- or two-time i.t. administration for cases in which surgical approaches are difficult to apply. Meanwhile, cancer patients can be free from the pain of multiple surgical treatments and have the advantage of therapy through a simple i.t. administration.
Collapse
Affiliation(s)
- Jang Il Kim
- Biomaterials Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | | | | | | | | | | |
Collapse
|
234
|
Liu C, Krishnan J, Xu XY. A systems-based mathematical modelling framework for investigating the effect of drugs on solid tumours. Theor Biol Med Model 2011; 8:45. [PMID: 22152406 PMCID: PMC3261817 DOI: 10.1186/1742-4682-8-45] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 12/07/2011] [Indexed: 11/23/2022] Open
Abstract
Background Elucidating the effects of drugs on solid tumours is a highly challenging multi-level problem, since this involves many complexities associated with transport and cellular response, which in turn is characterized by highly non-linear chemical signal transduction. Appropriate systems frameworks are needed to seriously address the sources of these complexities, especially from the cellular side. Results We develop a skeletal modelling framework incorporating interstitial drug transport, intracellular signal processing and cell population descriptions. The descriptions aim to appropriately capture the nature of information flow. The model is deliberately formulated to start with simple intracellular descriptions so that additional features can be incorporated in a modular fashion. Two kinds of intracellular signalling modules which describe the drug effect were considered, one a monostable switch and the other a bistable switch. Analysis of our model revealed how different drug stimuli can lead to cell killing in the tumour. Interestingly both modules considered exhibited similar trends. The effects of important parameters were also studied. Conclusions We have created a predictive systems platform integrating drug transport and cellular response which can be systematically augmented to include additional layers of cellular complexity. Our results indicate that intracellular signalling models which are qualitatively different can give rise to similar behaviour to simple (and typical) stimuli, and that validating intracellular descriptions must be performed with care by considering a variety of drug stimuli.
Collapse
Affiliation(s)
- Cong Liu
- Chemical Engineering and Chemical Technology, Imperial College London, South Kensington, London SW7 2AZ, UK
| | | | | |
Collapse
|
235
|
Cho H, Kwon GS. Polymeric micelles for neoadjuvant cancer therapy and tumor-primed optical imaging. ACS NANO 2011; 5:8721-8729. [PMID: 21999531 PMCID: PMC3879117 DOI: 10.1021/nn202676u] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Poly(ethylene glycol)-block-poly(d,l-lactic acid) (PEG-b-PLA) micelles act as a 3-in-1 nanocontainer for three poorly water-soluble drugs-paclitaxel, 17-allylamino-17-demethoxygeldanamycin, and rapamycin (PTX/17-AAG/RAPA)-for cancer therapy. In a LS180 human colon xenograft model, a single intravenous (IV) injection of 3-in-1 PEG-b-PLA micelles reduced tumor volume by 1.6-fold with <10% body weight change. In a second step, IV injection of poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) micelles carrying a carbocyanine dye (1,1'-dioctadecyl tetramethyl indotricarbocyanine iodide (DiR)) after 48 h resulted in a 2.1-fold higher near-infrared (NIR) optical signal from excised solid tumors versus a negative control, presumably due to a reduction in tumor cell density and interstitial tumor pressure. Thus, a tandem of 3-in-1 PEG-b-PLA and PEG-b-PCL micelles could potentially be used for neoadjuvant cancer therapy and tumor-primed NIR optical imaging for intraoperative surgical guidance in oncology, offering a promising multimodal strategy for cancer therapy and imaging.
Collapse
|
236
|
Li L, Guan Y, Liu H, Hao N, Liu T, Meng X, Fu C, Li Y, Qu Q, Zhang Y, Ji S, Chen L, Chen D, Tang F. Silica nanorattle-doxorubicin-anchored mesenchymal stem cells for tumor-tropic therapy. ACS NANO 2011; 5:7462-70. [PMID: 21854047 DOI: 10.1021/nn202399w] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Low targeting efficiency is one of the biggest limitations for nanoparticulate drug delivery system-based cancer therapy. In this study, an efficient approach for tumor-targeted drug delivery was developed with mesenchymal stem cells as the targeting vehicle and a silica nanorattle as the drug carrier. A silica nanorattle-doxorubicin drug delivery system was efficiently anchored to mesenchymal stem cells (MSCs) by specific antibody-antigen recognitions at the cytomembrane interface without any cell preconditioning. Up to 1500 nanoparticles were uploaded to each MSC cell with high cell viability and tumor-tropic ability. The intracellular retention time of the silica nanorattle was no less than 48 h, which is sufficient for cell-directed tumor-tropic delivery. In vivo experiments proved that the burdened MSCs can track down the U251 glioma tumor cells more efficiently and deliver doxorubicin with wider distribution and longer retention lifetime in tumor tissues compared with free DOX and silica nanorattle-encapsulated DOX. The increased and prolonged DOX intratumoral distribution further contributed to significantly enhanced tumor-cell apoptosis. This strategy has potential to be developed as a robust and generalizable method for targeted tumor therapy with high efficiency and low systematic toxicity.
Collapse
Affiliation(s)
- Linlin Li
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Kievit FM, Zhang M. Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H217-47. [PMID: 21842473 PMCID: PMC3397249 DOI: 10.1002/adma.201102313] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/12/2011] [Indexed: 05/03/2023]
Abstract
Cancer nanotheranostics aims to combine imaging and therapy of cancer through use of nanotechnology. The ability to engineer nanomaterials to interact with cancer cells at the molecular level can significantly improve the effectiveness and specificity of therapy to cancers that are currently difficult to treat. In particular, metastatic cancers, drug-resistant cancers, and cancer stem cells impose the greatest therapeutic challenge for targeted therapy. Targeted therapy can be achieved with appropriately designed drug delivery vehicles such as nanoparticles, adult stem cells, or T cells in immunotherapy. In this article, we first review the different types of nanotheranostic particles and their use in imaging, followed by the biological barriers they must bypass to reach the target cancer cells, including the blood, liver, kidneys, spleen, and particularly the blood-brain barrier. We then review how nanotheranostics can be used to improve targeted delivery and treatment of cancer cells. Finally, we discuss development of nanoparticles to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
238
|
Spugnini EP, Renaud SM, Buglioni S, Carocci F, Dragonetti E, Murace R, Cardelli P, Vincenzi B, Baldi A, Citro G. Electrochemotherapy with cisplatin enhances local control after surgical ablation of fibrosarcoma in cats: an approach to improve the therapeutic index of highly toxic chemotherapy drugs. J Transl Med 2011; 9:152. [PMID: 21917133 PMCID: PMC3182914 DOI: 10.1186/1479-5876-9-152] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 09/14/2011] [Indexed: 11/18/2022] Open
Abstract
Background Cancer is one of the most difficult current health challenges, being responsible for millions of deaths yearly. Systemic chemotherapy is the most common therapeutic approach, and the prevailing orientation calls for the administration of the maximum tolerated dose; however, considerable limitations exist including toxicities to healthy tissues and low achievable drug concentrations at tumor sites. Electrochemotherapy (ECT) is a tumor treatment that combines the systemic or local delivery of anticancer drugs with the application of permeabilizing electric pulses. In this article we evaluate the capability of ECT to allow the use of cisplatin despite its high toxicity in a spontaneous feline model of soft tissue sarcoma. Methods A cohort of sixty-four cats with incompletely excised sarcomas were treated with cisplatin-based adjuvant ECT and monitored for side effects. Their response was compared to that of fourteen cats treated with surgery alone. Results The toxicities were minimal and mostly treated symptomatically. ECT resulted in increased local control (median not reached at the time of writing) with a mean time to recurrence of 666 days versus 180 of controls. Conclusions We conclude that ECT is a safe and efficacious therapy for solid tumors; its use may be considered as part of strategies for the reintroduction of drugs with a narrow therapeutic index in the clinical protocols.
Collapse
|
239
|
Anti-MUC1 monoclonal antibody (C595) and docetaxel markedly reduce tumor burden and ascites, and prolong survival in an in vivo ovarian cancer model. PLoS One 2011; 6:e24405. [PMID: 21931707 PMCID: PMC3170300 DOI: 10.1371/journal.pone.0024405] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/09/2011] [Indexed: 12/31/2022] Open
Abstract
MUC1 is associated with cellular transformation and tumorigenicity and is considered as an important tumor-associated antigen (TAA) for cancer therapy. We previously reported that anti-MUC1 monoclonal antibody C595 (MAb C595) plus docetaxel (DTX) increased efficacy of DTX alone and caused cultured human epithelial ovarian cancer (EOC) cells to undergo apoptosis. To further study the mechanisms of this combination-mediated apoptosis, we investigated the effectiveness of this combination therapy in vivo in an intraperitoneal (i.p.) EOC mouse model. OVCAR-3 cells were implanted intraperitoneally in female athymic nude mice and allowed to grow tumor and ascites. Mice were then treated with single MAb C595, DTX, combination test (MAb C595 and DTX), combination control (negative MAb IgG3 and DTX) or vehicle control i.p for 3 weeks. Treated mice were killed 4 weeks post-treatment. Ascites volume, tumor weight, CA125 levels from ascites and survival of animals were assessed. The expression of MUC1, CD31, Ki-67, TUNEL and apoptotic proteins in tumor xenografts was evaluated by immunohistochemistry. MAb C595 alone inhibited i.p. tumor growth and ascites production in a dose-dependent manner but did not obviously prevent tumor development. However, combination test significantly reduced ascites volume, tumor growth and metastases, CA125 levels in ascites and improved survival of treated mice compared with single agent-treated mice, combination control or vehicle control-treated mice (P<0.05). The data was in a good agreement with that from cultured cells in vitro. The mechanisms behind the observed effects could be through targeting MUC1 antigens, inhibition of tumor angiogenesis, and induction of apoptosis. Our results suggest that this combination approach can effectively reduce tumor burden and ascites, prolong survival of animals through induction of tumor apoptosis and necrosis, and may provide a potential therapy for advanced metastatic EOC.
Collapse
|
240
|
Lovell JF, Huynh E, MacDonald TD, Lin Q, Zheng G. Research Highlights. Nanomedicine (Lond) 2011; 6:1155. [DOI: 10.2217/nnm.11.116] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Jonathan F Lovell
- Ontario Cancer Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, ON, M5G 1L7, Canada
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Elizabeth Huynh
- Ontario Cancer Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, ON, M5G 1L7, Canada
| | - Thomas D MacDonald
- Ontario Cancer Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Pharmaceutical Sciences, University of Toronto, ON, M5G 1L7, Canada
| | - Qiaoya Lin
- Department of Medical Biophysics, University of Toronto, ON, M5G 1L7, Canada
- Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | | |
Collapse
|
241
|
Abstract
Nanomedicine, the application of nanotechnology to medicine, enabled the development of nanoparticle therapeutic carriers. These drug carriers are passively targeted to tumors through the enhanced permeability and retention effect, so they are ideally suited for the delivery of chemotherapeutics in cancer treatment. Indeed, advances in nanomedicine have rapidly translated into clinical practice. To date, there are five clinically approved nanoparticle chemotherapeutics for cancer and many more under clinical investigation. In this review, we discuss the various nanoparticle drug delivery platforms and the important concepts involved in nanoparticle drug delivery. We also review the clinical data on the approved nanoparticle therapeutics as well as the nanotherapeutics under clinical investigation.
Collapse
Affiliation(s)
- Andrew Z Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | | | |
Collapse
|
242
|
Choo EF, Alicke B, Boggs J, Dinkel V, Gould S, Grina J, West K, Menghrajani K, Ran Y, Rudolph J, Wenglowsky S. Preclinical assessment of novel BRAF inhibitors: integrating pharmacokinetic-pharmacodynamic modelling in the drug discovery process. Xenobiotica 2011; 41:1076-87. [DOI: 10.3109/00498254.2011.603384] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
243
|
ANDHARIYA NIDHI, CHUDASAMA BHUPENDRA, UPADHYAY RV, MEHTA RV. METHYLENE BLUE LOADED SILICA ENCAPSULATED MAGNETITE NANOPARTICLES: A POTENTIAL DRUG DELIVERY VECTOR FOR PHOTODYNAMIC THERAPY. INTERNATIONAL JOURNAL OF NANOSCIENCE 2011. [DOI: 10.1142/s0219581x11008472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this article, we describe synthesis of a novel drug delivery vector (DDV) for photodynamic therapy (PDT). The DDV consists of a magnetite core surrounded by a thin layer of functionalized silica. These core–shell structures are loaded with a photosensitizer (PS) drug "Methylene Blue" (MB). Magnetite nanostructures are produce by the well-established chemical co-precipitation technique and encapsulated in silica shell by modified process of hydrolysis and condensation of tetraethyl orthosilicate (TEOS). MB is grafted into the pores of silica shell by demethylation reaction. Reaction kinetics has been established for tunable loading of PS in DDV. Physical and chemical properties of composite nanostructures are determined by X-ray diffraction (XRD), dynamic light scattering (DLS), transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIR) and vibrating sample magnetometry (VSM). Amount of PS loading in DDV is measured by UV-Visible spectroscopy. Smaller size, biocompatibility, tunable loading of PS and capabilities of magnetic guidance, makes this DDV, a potential candidate for the treatment of malignant tumors by PDT.
Collapse
Affiliation(s)
- NIDHI ANDHARIYA
- Department of Physics, Bhavnagar University, Bhavnagar, Gujarat 364022, India
| | - BHUPENDRA CHUDASAMA
- School of Physics and Materials Science, Thapar University, Patiala, Punjab 147004, India
| | - R. V. UPADHYAY
- P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Gujarat 364022, India
| | - R. V. MEHTA
- Department of Physics, Bhavnagar University, Bhavnagar, Gujarat 364022, India
| |
Collapse
|
244
|
Llevot A, Astruc D. Applications of vectorized gold nanoparticles to the diagnosis and therapy of cancer. Chem Soc Rev 2011; 41:242-57. [PMID: 21785769 DOI: 10.1039/c1cs15080d] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This critical review focuses on the anti-cancer fight using gold nanoparticles (AuNPs) functionalized with chemotherapeutic drugs in so-called "complexes" (supramolecular assemblies) and "conjugates" (covalent assemblies) as vectors. There is a considerable body of recent literature on various tumor-imaging techniques using the surface plasmon band (SPB) and the "passive" and "active" vectorization of anti-cancer drugs. This article reviews the main concepts and the most recent literature data with emphasis on AuNP preparation, cytotoxicities and use in selective targeting of cancer cells with over-expressed receptors for diagnosis and therapy (108 references).
Collapse
|
245
|
Narang AS, Varia S. Role of tumor vascular architecture in drug delivery. Adv Drug Deliv Rev 2011; 63:640-58. [PMID: 21514334 DOI: 10.1016/j.addr.2011.04.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/23/2011] [Accepted: 04/05/2011] [Indexed: 12/14/2022]
Abstract
Tumor targeted drug delivery has the potential to improve cancer care by reducing non-target toxicities and increasing the efficacy of a drug. Tumor targeted delivery of a drug from the systemic circulation, however, requires a thorough understanding of tumor pathophysiology. A growing or receding (under the impact of therapy) tumor represents a dynamic environment with changes in its angiogenic status, cell mass, and extracellular matrix composition. An appreciation of the salient characteristics of tumor vascular architecture and the unique biochemical markers that may be used for targeting drug therapy is important to overcome barriers to tumor drug therapy and to facilitate targeted drug delivery. This review discusses the unique aspects of tumor vascular architecture that need to be overcome or exploited for tumor targeted drug delivery.
Collapse
Affiliation(s)
- Ajit S Narang
- Bristol-Myers Squibb, Co., One Squibb Dr., PO Box 191, New Brunswick, NJ 08903-0191, USA.
| | | |
Collapse
|
246
|
Attaluri A, Ma R, Qiu Y, Li W, Zhu L. Nanoparticle distribution and temperature elevations in prostatic tumours in mice during magnetic nanoparticle hyperthermia. Int J Hyperthermia 2011; 27:491-502. [DOI: 10.3109/02656736.2011.584856] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
247
|
Di Fede G, Bronte G, Rizzo S, Rolfo Cervetto C, Cocorullo G, Gulotta G, Bazan V, Russo A. Monoclonal antibodies and antibody fragments: state of the art and future perspectives in the treatment of non-haematological tumors. Expert Opin Biol Ther 2011; 11:1433-45. [DOI: 10.1517/14712598.2011.594436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
248
|
Wong HL, Shen Z, Lu Z, Wientjes MG, Au JLS. Paclitaxel tumor-priming enhances siRNA delivery and transfection in 3-dimensional tumor cultures. Mol Pharm 2011; 8:833-40. [PMID: 21417439 PMCID: PMC3109178 DOI: 10.1021/mp1004383] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The clinical development of siRNA cancer therapeutics is limited by the poor interstitial transport and inefficient transfection in solid tumors. We have shown that paclitaxel pretreatment, by inducing apoptosis, causes expansion of the interstitial space and thereby improves nanoparticle delivery and transport in tumor interstitium (referred to as paclitaxel tumor priming) and efficacy of nanomedicines in tumor-bearing animals. The present study evaluated whether paclitaxel tumor priming improves the delivery and transfection of siRNA in 2- and 3-dimensional cultures of human oropharyngeal carcinoma FaDu cells. We used the fluorescent siGLO and confocal microcopy to monitor transport, and used survivin siRNA and immunostaining and immunoblotting to monitor transfection. Survivin is a chemoresistance gene/protein, inducible by chemotherapy. siRNA was loaded in cationic liposomes. The results showed that pretreatment with 50-200 nM paclitaxel (24 or 48 h before siRNA) enhanced the total uptake of siGLO into monolayers (∼15%, p < 0.05), and the depth of penetration into 3-dimensional spheroids and tumor fragment histocultures (2.1- to 2.5-times greater area under the penetration-depth curve). In both monolayer cells and histocultures, paclitaxel pretreatment induced survivin upregulation (p < 0.05). Survivin siRNA alone decreased the survivin levels in a dose-dependent manner, and applying survivin siRNA after paclitaxel pretreatment completely abolished the paclitaxel-induced survivin increases. These findings indicate that paclitaxel tumor priming did not compromise the siRNA functionality. In summary, paclitaxel tumor priming improved the penetration, transfection and functionality of siRNA in tumors, thus offering a promising and practical means to develop chemo-siRNA cancer gene therapy.
Collapse
Affiliation(s)
- Ho Lun Wong
- College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210, USA
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Zancong Shen
- College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210, USA
| | - Ze Lu
- Optimum Therapeutics LLC, OSU Science Tech Village, Columbus, OH 43212, USA
| | - M. Guillaume Wientjes
- College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210, USA
| | - Jessie L.-S. Au
- College of Pharmacy, The Ohio State University, 500 West 12 Avenue, Columbus, OH 43210, USA
| |
Collapse
|
249
|
Abstract
A mathematical model of interstitial fluid flow is developed, based on the application of the governing equations for fluid flow, i.e., the conservation laws for mass and momentum, to physiological systems containing solid tumors. The discretized form of the governing equations, with appropriate boundary conditions, is developed for a predefined tumor geometry. The interstitial fluid pressure and velocity are calculated using a numerical method, element based finite volume. Simulations of interstitial fluid transport in a homogeneous solid tumor demonstrate that, in a uniformly perfused tumor, i.e., one with no necrotic region, because of the interstitial pressure distribution, the distribution of drug particles is non-uniform. Pressure distribution for different values of necrotic radii is examined and two new parameters, the critical tumor radius and critical necrotic radius, are defined. Simulation results show that: 1) tumor radii have a critical size. Below this size, the maximum interstitial fluid pressure is less than what is generally considered to be effective pressure (a parameter determined by vascular pressure, plasma osmotic pressure, and interstitial osmotic pressure). Above this size, the maximum interstitial fluid pressure is equal to effective pressure. As a consequence, drugs transport to the center of smaller tumors is much easier than transport to the center of a tumor whose radius is greater than the critical tumor radius; 2) there is a critical necrotic radius, below which the interstitial fluid pressure at the tumor center is at its maximum value. If the tumor radius is greater than the critical tumor radius, this maximum pressure is equal to effective pressure. Above this critical necrotic radius, the interstitial fluid pressure at the tumor center is below effective pressure. In specific ranges of these critical sizes, drug amount and therefore therapeutic effects are higher because the opposing force, interstitial fluid pressure, is low in these ranges.
Collapse
Affiliation(s)
- M. Soltani
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - P. Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
- * E-mail:
| |
Collapse
|
250
|
Xu J, Ganesh S, Amiji M. Non-condensing polymeric nanoparticles for targeted gene and siRNA delivery. Int J Pharm 2011; 427:21-34. [PMID: 21621597 DOI: 10.1016/j.ijpharm.2011.05.036] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/11/2011] [Accepted: 05/12/2011] [Indexed: 12/27/2022]
Abstract
Gene therapy has shown a tremendous potential to benefit patients in a variety of disease conditions. However, finding a safe and effective systemic delivery system is the major obstacle in this area. Although viral vectors showed promise for high transfection rate, the immunogenicity associated with these systems has hindered further development. As an alternative to viral gene delivery, this review focuses on application of novel safe and effective non-condensing polymeric systems that have shown high transgene expression when administered systemically or by the oral route. Type B gelatin-based engineered nanocarriers were evaluated for passive and active tumor-targeted delivery and transfection using both reporter and therapeutic plasmid DNA. Additionally, we have shown that nanoparticles-in-microsphere oral system (NiMOS) can efficiently deliver reporter and therapeutic gene constructs in the gastrointestinal tract. Additionally, there has been a significant recent interest in the use small interfering RNA (siRNA) as a therapeutic system for gene silencing. Both gelatin nanoparticles and NiMOS have shown activity in systemic and oral delivery of siRNA, respectively.
Collapse
Affiliation(s)
- Jing Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 110 Mugar Life Sciences Building, 360 Huntington Avenue, Boston, MA 02115, United States
| | | | | |
Collapse
|