301
|
Homan K, Shah J, Gomez S, Gensler H, Karpiouk A, Brannon-Peppas L, Emelianov S. Silver nanosystems for photoacoustic imaging and image-guided therapy. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:021316. [PMID: 20459238 PMCID: PMC2859084 DOI: 10.1117/1.3365937] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 01/05/2010] [Accepted: 01/06/2010] [Indexed: 05/23/2023]
Abstract
Due to their optical absorption properties, metallic nanoparticles are excellent photoacoustic imaging contrast agents. A silver nanosystem is presented here as a potential contrast agent for photoacoustic imaging and image-guided therapy. Currently, the nanosystem consists of a porous silver layer deposited on the surface of spherical silica cores ranging in diameter from 180 to 520 nm. The porous nature of the silver layer will allow for release of drugs or other therapeutic agents encapsulated in the core in future applications. In their current PEGylated form, the silver nanosystem is shown to be nontoxic in vitro at concentrations of silver up to 2 mgml. Furthermore, the near-infrared absorbance properties of the nanosystem are demonstrated by measuring strong, concentration-dependent photoacoustic signal from the silver nanosystem embedded in an ex vivo tissue sample. Our study suggests that silver nanosystems can be used as multifunctional agents capable of augmenting image-guided therapy techniques.
Collapse
Affiliation(s)
- Kimberly Homan
- The University of Texas at Austin, Biomedical Engineering Department, 1 University Station C0800, Austin, Texas 78712, USA
| | | | | | | | | | | | | |
Collapse
|
302
|
The inhibitory effects of rh-endostatin (YH-16) in combination with radiotherapy on lung adenocarcinoma A549 in mice and the underlying mechanisms. ACTA ACUST UNITED AC 2010; 30:108-12. [DOI: 10.1007/s11596-010-0120-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Indexed: 11/24/2022]
|
303
|
Modulating the pharmacokinetics of therapeutic antibodies. Biotechnol Lett 2010; 32:609-22. [PMID: 20131077 DOI: 10.1007/s10529-010-0214-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 01/06/2010] [Accepted: 01/09/2010] [Indexed: 12/22/2022]
Abstract
With the advent of antibody fragments and alternative binding scaffolds, that are devoid of Fc-regions, strategies to increase the half-life of small proteins are becoming increasingly important. Currently, the established method is chemical PEGylation, but more elaborate approaches are being described such as polysialylation, amino acid polymers and albumin-binding derivatives. This article reviews the main strategies for pharmacokinetic enhancement, primarily chemical conjugates and recombinant fusions that increase apparent molecular weight or hydrodynamic radius or interact with serum albumin which itself has a long plasma half-life. We highlight the key chemical linkage methods that preserve antibody function and retain stability and look forward to the next generation of technologies which promise to make better quality pharmaceuticals with lower side effects. Although restricted to antibodies, all of the approaches covered can be applied to other biotherapeutics.
Collapse
|
304
|
Zhang Y, Hansen JK, Xiang L, Kawa S, Onda M, Ho M, Hassan R, Pastan I. A flow cytometry method to quantitate internalized immunotoxins shows that taxol synergistically increases cellular immunotoxins uptake. Cancer Res 2010; 70:1082-9. [PMID: 20103626 DOI: 10.1158/0008-5472.can-09-2405] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor microenvironments present significant barriers to penetration by antibodies, immunoconjugates, and other immunotoxins. In this report, we illustrate a novel strategy to increase tumor cell uptake of immunotoxin by combination with Taxol. SS1P is an immunotoxin composed of the Fv portion of a mesothelin-specific antibody fused to a bacterial toxin that is presently undergoing phase II testing in mesothelioma. Using novel flow cytometry and gel filtration methods, we quantified SS1P uptake in individual tumor cells along with levels of shed mesothelin (sMSLN), a barrier of SS1P therapy. The validity of our flow cytometric method was confirmed by the ability to similarly quantitate tumor cell uptake of Herceptin and an immunotoxin targeting HER2/neu. SS1P uptake peaked several hours after SS1P was cleared from the blood, reflecting an intratumor distribution process of SS1P that is independent of blood supply. Using the methods developed, we demonstrated that Taxol could improve SS1P penetration into tumors in parallel with an associated reduction of sMSLN in tumor extracellular fluid. Our findings offer a mechanistic rationale to combine SS1P with Taxol or another cytotoxic drug as a strategy to increase immunotoxin uptake by tumor cells. Further, we suggest one basis to understand why chemotherapy and antibody-based therapies cooperate when combined in cancer treatment.
Collapse
Affiliation(s)
- Yujian Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-4264, USA
| | | | | | | | | | | | | | | |
Collapse
|
305
|
Ferrari M. Frontiers in cancer nanomedicine: directing mass transport through biological barriers. Trends Biotechnol 2010; 28:181-8. [PMID: 20079548 DOI: 10.1016/j.tibtech.2009.12.007] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 12/16/2009] [Accepted: 12/17/2009] [Indexed: 01/30/2023]
Abstract
The physics of mass transport within body compartments and across biological barriers differentiates cancers from healthy tissues. Variants of nanoparticles can be manufactured in combinatorially large sets, varying by only one transport-affecting design parameter at a time. Nanoparticles can also be used as building blocks for systems that perform sequences of coordinated actions, in accordance with a prescribed logic. We refer to these as Logic-Embedded Vectors (LEVs). Nanoparticles and LEVs are ideal probes for the determination of mass transport laws in tumors, acting as imaging contrast enhancers, and can be employed for lesion-selective delivery of therapy. Their size, shape, density and surface chemistry dominate convective transport in the bloodstream, margination, cell adhesion, selective cellular uptake, as well as sub-cellular trafficking and localization. As argued here, the understanding of transport differentials in cancer, termed 'transport oncophysics', reveals a promising new frontier in oncology: the development of lesion-specific delivery particulates that exploit mass transport differentials to deploy treatment of greater efficacy and reduced side effects.
Collapse
Affiliation(s)
- Mauro Ferrari
- University of Texas Health Science Center at Houston (UTHSC-H), Department of NanoMedicine and Biomedical Engineering, 1825 Pressler, Suite 537, Houston, TX 77030, USA.
| |
Collapse
|
306
|
Abstract
The paradigm of using nanoparticulate pharmaceutical carriers has been well established over the past decade, both in pharmaceutical research and in the clinical setting. Drug carriers are expected to stay in the blood for long time, accumulate in pathological sites with affected and leaky vasculature (tumors, inflammations, and infarcted areas) via the enhanced permeability and retention (EPR) effect, and facilitate targeted delivery of specific ligand-modified drugs and drug carriers into poorly accessible areas. Among various approaches to specifically target drug-loaded carrier systems to required pathological sites in the body, two seem to be most advanced--passive (EPR effect-mediated) targeting, based on the longevity of the pharmaceutical carrier in the blood and its accumulation in pathological sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the surface of pharmaceutical carriers to recognize and bind pathological cells. Here, we will consider and discuss these two targeting approaches using tumor targeting as an example.
Collapse
|
307
|
Godin B, Driessen WHP, Proneth B, Lee SY, Srinivasan S, Rumbaut R, Arap W, Pasqualini R, Ferrari M, Decuzzi P. An integrated approach for the rational design of nanovectors for biomedical imaging and therapy. ADVANCES IN GENETICS 2010; 69:31-64. [PMID: 20807601 DOI: 10.1016/s0065-2660(10)69009-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of nanoparticles for the early detection, cure, and imaging of diseases has been proved already to have a colossal potential in different biomedical fields, such as oncology and cardiology. A broad spectrum of nanoparticles are currently under development, exhibiting differences in (i) size, ranging from few tens of nanometers to few microns; (ii) shape, from the classical spherical beads to discoidal, hemispherical, cylindrical, and conical; (iii) surface functionalization, with a wide range of electrostatic charges and biomolecule conjugations. Clearly, the library of nanoparticles generated by combining all possible sizes, shapes, and surface physicochemical properties is enormous. With such a complex scenario, an integrated approach is here proposed and described for the rational design of nanoparticle systems (nanovectors) for the intravascular delivery of therapeutic and imaging contrast agents. The proposed integrated approach combines multiscale/multiphysics mathematical models with in vitro assays and in vivo intravital microscopy (IVM) experiments and aims at identifying the optimal combination of size, shape, and surface properties that maximize the nanovectors localization within the diseased microvasculature.
Collapse
Affiliation(s)
- Biana Godin
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
308
|
Abstract
The inhibition of angiogenesis represents a major step toward a more selective and better-tolerated therapy of cancer. An alternative way to take advantage of a tumor's absolute dependence on a functional neovasculature is illustrated by the strategy of "antibody-based vascular tumor targeting." This technology aims at the selective delivery of bioactive molecules to the tumor site by their conjugation to a carrier antibody reactive with a tumor-associated vascular antigen. A number of high-affinity monoclonal antibodies are nowadays available which have demonstrated a remarkable ability to selectively localize to the tumor vasculature. Indeed, some of them have already progressed from preclinical animal experiments to clinical studies in patients with cancer, acting as vehicles for the site-specific pharmacodelivery of proinflammatory cytokines or radionuclides.In this chapter, we present a selection of well-characterized markers of angiogenesis which have proven to be suitable targets for antibody-based vascular targeting approaches. Furthermore, different transcriptomic and proteomic methodologies for the discovery of novel vascular tumor markers are described. In the last two sections, we focus on the discussion of antibody-based vascular tumor targeting strategies for imaging and therapy applications in oncology.
Collapse
Affiliation(s)
- Christoph Schliemann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zürich, Zürich, Switzerland
| | | |
Collapse
|
309
|
Cooperative nanomaterial system to sensitize, target, and treat tumors. Proc Natl Acad Sci U S A 2009; 107:981-6. [PMID: 20080556 DOI: 10.1073/pnas.0909565107] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A significant barrier to the clinical translation of systemically administered therapeutic nanoparticles is their tendency to be removed from circulation by the mononuclear phagocyte system. The addition of a targeting ligand that selectively interacts with cancer cells can improve the therapeutic efficacy of nanomaterials, although these systems have met with only limited success. Here, we present a cooperative nanosystem consisting of two discrete nanomaterials. The first component is gold nanorod (NR) "activators" that populate the porous tumor vessels and act as photothermal antennas to specify tumor heating via remote near-infrared laser irradiation. We find that local tumor heating accelerates the recruitment of the second component: a targeted nanoparticle consisting of either magnetic nanoworms (NW) or doxorubicin-loaded liposomes (LP). The targeting species employed in this work is a cyclic nine-amino acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-Arg-Gly-Cys) that binds to the stress-related protein, p32, which we find to be upregulated on the surface of tumor-associated cells upon thermal treatment. Mice containing xenografted MDA-MB-435 tumors that are treated with the combined NR/LyP-1LP therapeutic system display significant reductions in tumor volume compared with individual nanoparticles or untargeted cooperative system.
Collapse
|
310
|
Han B, Teo KY. Effects of freezing on intratumoral drug transport. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2009; 2009:246-9. [PMID: 19964211 DOI: 10.1109/iembs.2009.5333804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Efficacy of many novel therapeutic agents are impaired by hindered interstitial diffusion in tumor. In the context of overcoming this drug delivery barrier, a hypothesis was postulated that freeze/thaw (F/T) may induce favorable changes of tumor tissue microstructure to facilitate the interstitial diffusion. This hypothesis may also be relevant to develop a mechanistically derived chemotherapeutic strategy for cryo-treated tumors. In the present study, this hypothesis was tested by characterizing the effects of F/T on the interstitial diffusion using an in vitro engineered tumor model (ET). The diffusion coefficients of FITC-labeled dextran was measured within the frozen/thawed and unfrozen ETs. The results showed that the diffusion coefficients increased after F/T but the extent of increase was dependent on the size of dextran. This implies that the combination of cryosurgery and chemotherapy should be designed considering the biophysical changes of tissues after freeze/thaw and the diffusion characteristics of drug molecules.
Collapse
Affiliation(s)
- Bumsoo Han
- University of Texas at Arlington, Arlington, TX 76019, USA.
| | | |
Collapse
|
311
|
Krishna ADS, Mandraju RK, Kishore G, Kondapi AK. An efficient targeted drug delivery through apotransferrin loaded nanoparticles. PLoS One 2009; 4:e7240. [PMID: 19806207 PMCID: PMC2752169 DOI: 10.1371/journal.pone.0007240] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 08/31/2009] [Indexed: 02/07/2023] Open
Abstract
Background Cancerous state is a highly stimulated environment of metabolically active cells. The cells under these conditions over express selective receptors for assimilation of factors essential for growth and transformation. Such receptors would serve as potential targets for the specific ligand mediated transport of pharmaceutically active molecules. The present study demonstrates the specificity and efficacy of protein nanoparticle of apotransferrin for targeted delivery of doxorubicin. Methodology/Principal Findings Apotransferrin nanoparticles were developed by sol-oil chemistry. A comparative analysis of efficiency of drug delivery in conjugated and non-conjugated forms of doxorubicin to apotransferrin nanoparticle is presented. The spherical shaped apotransferrin nanoparticles (nano) have diameters of 25–50 ηm, which increase to 60–80 ηm upon direct loading of drug (direct-nano), and showed further increase in dimension (75–95 ηm) in conjugated nanoparticles (conj-nano). The competitive experiments with the transferrin receptor specific antibody showed the entry of both conj-nano and direct-nano into the cells through transferrin receptor mediated endocytosis. Results of various studies conducted clearly establish the superiority of the direct-nano over conj-nano viz. (a) localization studies showed complete release of drug very early, even as early as 30 min after treatment, with the drug localizing in the target organelle (nucleus) (b) pharmacokinetic studies showed enhanced drug concentrations, in circulation with sustainable half-life (c) the studies also demonstrated efficient drug delivery, and an enhanced inhibition of proliferation in cancer cells. Tissue distribution analysis showed intravenous administration of direct nano lead to higher drug localization in liver, and blood as compared to relatively lesser localization in heart, kidney and spleen. Experiments using rat cancer model confirmed the efficacy of the formulation in regression of hepatocellular carcinoma with negligible toxicity to kidney and liver. Conclusions The present study thus demonstrates that the direct-nano is highly efficacious in delivery of drug in a target specific manner with lower toxicity to heart, liver and kidney.
Collapse
Affiliation(s)
| | | | - Golla Kishore
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
| | - Anand Kumar Kondapi
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
- * E-mail:
| |
Collapse
|
312
|
Lin WZ, Li ZF, Tsang SY, Lung LKW, Wang DK, Chan WY, Zhu YK, Lee SST, Cheung WT. Identification and characterization of a novel CXC chemokine in xenograft tumor induced by mas-overexpressing cells. Int J Cancer 2009; 125:1316-27. [PMID: 19408311 DOI: 10.1002/ijc.24440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Overexpressions of G protein-coupled receptor (GPCR) with elevated downstream signaling events have been reported in various tumors. However, the cellular mechanism that GPCR overexpression leads to tumor formation is largely unknown. The orphan GPCR mas was originally isolated from a human epidermoid carcinoma. In vivo studies of mas-overexpressing cells suggested that xenograft tumor formation was positively correlated with the levels of mas expression. Histochemical analysis indicated that xenograft tumor consisted of mas-transfected and stromal cells. Biochemical analyses revealed that cells overexpressing mas exhibited significantly increased anchorage-independent growth, whereas there was no significant difference in cell proliferation in comparison with empty vector-transfected control cells. Expression profiling using mRNA differential display and Northern analysis indicated an elevated expression of GRO and a novel CXC chemokines, tumor-induced factor (TIF), in mas-transfected cells and xenograft tumor. Bacterially expressed recombinant TIF was found to act as a neutrophil chemoattractant in a chemotactic assay. These results suggest that mas overexpression enables anchorage-independent growth of transformed cells, and interplays of secreted chemokines with stromal cells modulate xenograft tumor formation. Importantly, a novel CXC chemokine, TIF, was identified in the xenograft tumor tissues.
Collapse
Affiliation(s)
- Wen-Zhen Lin
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
313
|
Salloum M, Ma R, Weeks D, Zhu L. Controlling nanoparticle delivery in magnetic nanoparticle hyperthermia for cancer treatment: Experimental study in agarose gel. Int J Hyperthermia 2009; 24:337-45. [DOI: 10.1080/02656730801907937] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
314
|
Chen T, McIntosh D, He Y, Kim J, Tirrell DA, Scherrer P, Fenske DB, Sandhu AP, Cullis PR. Alkylated derivatives of poly(ethylacrylic acid) can be inserted into preformed liposomes and trigger pH-dependent intracellular delivery of liposomal contents. Mol Membr Biol 2009; 21:385-93. [PMID: 15764368 DOI: 10.1080/09687860400010516] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Poly(ethylacrylic acid) (PEAA) is a pH-sensitive polymer that undergoes a transition from a hydrophilic to a hydrophobic form as the pH is lowered from neutral to acidic values. In this work we show that pH sensitive liposomes capable of intracellular delivery can be constructed by inserting a lipid derivative of PEAA into preformed large unilamellar vesicles (LUV) using a simple one step incubation procedure. The lipid derivatives of PEAA were synthesized by reacting a small proportion (3%) of the carboxylic groups of PEAA with C10 alkylamines to produce C10-PEAA. Incubation of C10-PEAA with preformed LUV resulted in the association of up to 8% by weight of derivatized polymer with the LUV without inducing aggregation. The resulting C10-PEAA-LUV exhibited pH-dependent fusion and leakage of LUV contents on reduction of the external pH below pH 6.0 as demonstrated by lipid mixing and release of calcein encapsulated in the LUV. In addition, C10-PEAA-LUV exhibited pH dependent intracellular delivery properties following uptake into COS-7 cells with appreciable delivery to the cell cytoplasm as evidenced by the appearance of diffuse intracellular calcein fluorescence. It is demonstrated that the cytoplasmic delivery of calcein by C10-PEAA-LUV could be inhibited by agents (bafilomycin or chloroquine) that inhibit acidification of endosomal compartments, indicating that this intracellular delivery resulted from the pH-dependent destabilization of LUV and endosomal membranes by the PEAA component of the C10-PEAA-LUV. It is concluded that C10-PEAA-LUV represents a promising intracellular delivery system for in vitro and in vivo applications.
Collapse
Affiliation(s)
- Tao Chen
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Affiliation(s)
- Peter Olson
- Comprehensive Cancer Center, Diabetes Center and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143-0534, USA.
| | | |
Collapse
|
316
|
Lee SY, Ferrari M, Decuzzi P. Design of bio-mimetic particles with enhanced vascular interaction. J Biomech 2009; 42:1885-90. [PMID: 19523635 DOI: 10.1016/j.jbiomech.2009.05.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 05/12/2009] [Accepted: 05/12/2009] [Indexed: 11/25/2022]
Abstract
The majority of particle-based delivery systems for the 'smart' administration of therapeutic and imaging agents have a spherical shape, are made by polymeric or lipid materials, have a size in the order of few hundreds of nanometers and a negligibly small relative density to aqueous solutions. In the microcirculation and deep airways of the lungs, where the creeping flow assumption holds, such small spheres move by following the flow stream lines and are not affected by external volume force fields. A delivery system should be designed to drift across the stream lines and interact repeatedly with the vessel walls, so that vascular interaction could be enhanced. The numerical approach presented in [Gavze, E., Shapiro, M., 1997. Particles in a shear flow near a solid wall: effect of nonsphericity on forces and velocities. International Journal of Multiphase Flow 23, 155-182.] is, here, proposed as a tool to analyze the dynamics of arbitrarily shaped particles in a creeping flow, and has been extended to include the contribution of external force fields. As an example, ellipsoidal particles with aspect ratio 0.5 are considered. In the absence of external volume forces, a net lateral drift (margination) of the particles has been observed for Stokes number larger than unity (St>1); whereas, for smaller St, the particles oscillate with no net lateral motion. Under these conditions, margination is governed solely by particle inertia (geometry and particle-to-fluid density ratio). In the presence of volume forces, even for fairly small St, margination is observed but in a direction dictated by the external force field. It is concluded that a fine balance between size, shape and density can lead to EVI particles (particles with enhanced vascular interaction) that are able to sense endothelial cells for biological and biophysical abnormalities, mimicking circulating platelets and leukocytes.
Collapse
Affiliation(s)
- Sei-Young Lee
- Department of Biomedical Engineering, Center of Nanomedicine, University of Texas Health Science Center at Houston, TX, USA
| | | | | |
Collapse
|
317
|
Phan JH, Moffitt RA, Stokes TH, Liu J, Young AN, Nie S, Wang MD. Convergence of biomarkers, bioinformatics and nanotechnology for individualized cancer treatment. Trends Biotechnol 2009; 27:350-8. [PMID: 19409634 PMCID: PMC3779321 DOI: 10.1016/j.tibtech.2009.02.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Revised: 02/12/2009] [Accepted: 02/25/2009] [Indexed: 12/23/2022]
Abstract
Recent advances in biomarker discovery, biocomputing and nanotechnology have raised new opportunities in the emerging fields of personalized medicine (in which disease detection, diagnosis and therapy are tailored to each individual's molecular profile) and predictive medicine (in which genetic and molecular information is used to predict disease development, progression and clinical outcome). Here, we discuss advanced biocomputing tools for cancer biomarker discovery and multiplexed nanoparticle probes for cancer biomarker profiling, in addition to the prospects for and challenges involved in correlating biomolecular signatures with clinical outcome. This bio-nano-info convergence holds great promise for molecular diagnosis and individualized therapy of cancer and other human diseases.
Collapse
Affiliation(s)
- John H. Phan
- Departments of Biomedical Engineering and Electrical and Computer Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Building 4106, Atlanta, GA 30332, USA
| | - Richard A. Moffitt
- Departments of Biomedical Engineering and Electrical and Computer Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Building 4106, Atlanta, GA 30332, USA
| | - Todd H. Stokes
- Departments of Biomedical Engineering and Electrical and Computer Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Building 4106, Atlanta, GA 30332, USA
| | - Jian Liu
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle Suite 2001, Atlanta, GA 30322, USA
| | - Andrew N. Young
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine and the Grady Memorial Hospital, Atlanta, GA 30322, USA
| | - Shuming Nie
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle Suite 2001, Atlanta, GA 30322, USA
| | - May D. Wang
- Departments of Biomedical Engineering and Electrical and Computer Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Building 4106, Atlanta, GA 30332, USA
| |
Collapse
|
318
|
Juliano R, Bauman J, Kang H, Ming X. Biological barriers to therapy with antisense and siRNA oligonucleotides. Mol Pharm 2009; 6:686-95. [PMID: 19397332 PMCID: PMC2758224 DOI: 10.1021/mp900093r] [Citation(s) in RCA: 227] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Attaining the full therapeutic utility of antisense and siRNA oligonucleotides will require understanding of the biological barriers that stand between initial administration of these drugs and their final actions within cells. This review examines some of the key barriers that affect the biodistribution of oligonucleotides both in molecular form and when they are associated with nanocarriers. An understanding of the biological processes underlying these barriers will aid in the design of more effective delivery systems.
Collapse
Affiliation(s)
- R Juliano
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | |
Collapse
|
319
|
Annapragada A, Bellamkonda RV. Image-based determination of ‘physiomarkers’ for personalized cancer therapy. Future Oncol 2009; 5:409-11. [DOI: 10.2217/fon.09.21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ananth Annapragada
- School of Health Information Sciences, University of Texas at Houston, Houston, TX, USA
| | - Ravi V Bellamkonda
- Neurological Biomaterials & Cancer Therapeutics, Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 3108 UA Whitaker Bldg, 313 Ferst Drive, Atlanta, GA 30332-0535, USA
| |
Collapse
|
320
|
Perrault SD, Walkey C, Jennings T, Fischer HC, Chan WCW. Mediating tumor targeting efficiency of nanoparticles through design. NANO LETTERS 2009; 9:1909-15. [PMID: 19344179 DOI: 10.1021/nl900031y] [Citation(s) in RCA: 1103] [Impact Index Per Article: 68.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Here we systematically examined the effect of nanoparticle size (10-100 nm) and surface chemistry (i.e., poly(ethylene glycol)) on passive targeting of tumors in vivo. We found that the physical and chemical properties of the nanoparticles influenced their pharmacokinetic behavior, which ultimately determined their tumor accumulation capacity. Interestingly, the permeation of nanoparticles within the tumor is highly dependent on the overall size of the nanoparticle, where larger nanoparticles appear to stay near the vasculature while smaller nanoparticles rapidly diffuse throughout the tumor matrix. Our results provide design parameters for engineering nanoparticles for optimized tumor targeting of contrast agents and therapeutics.
Collapse
Affiliation(s)
- Steven D Perrault
- Institute of Biomaterials and Biomedical Engineering, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | | | | | | | | |
Collapse
|
321
|
Selective targeting of nanocarriers to neutrophils and monocytes. Ann Biomed Eng 2009; 37:1984-92. [PMID: 19387833 DOI: 10.1007/s10439-009-9702-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 04/15/2009] [Indexed: 12/23/2022]
Abstract
We previously identified and characterized cell-type selective binding peptides from random peptide phage display libraries. Here, we used one of these peptides (GGP) to target liposomal nanocarriers to leukocyte subsets. To profile the binding selectivity of GGP-coated liposomes to human blood cells, we performed flow cytometric analysis with whole anti-coagulated blood. It is shown that when liposomal nanocarriers present these peptides on their surface, they facilitated cell-type specific targeting of liposomes to neutrophils and monocytes in contrast to nontargeted liposomes. Our data suggest that engineering the appropriate number of targeting peptide ligands on the nanocarrier surface is a factor in cell-binding selectivity, as is dose. Increasing the peptide density on the surface of the liposomes from 250 to 500 molecules resulted in more binding to neutrophils and monocytes. Fluorescence confocal microscopy corroborated the flow cytometry data revealing that liposomes coated with targeting GGP peptides decorated the surface of targeting cells and facilitate cell uptake of payload as evidenced by nuclear localization of tracer. These data suggest that small peptides identified by phage display techniques can be used to target nanocarriers that potentially carry therapeutic or imaging agents to leukocyte subsets. This ability has important implications for diseases where neutrophils and monocytes play a major role such as arthritis, inflammatory bowel disease, chronic obstructive pulmonary disease, and glomerulonephritis.
Collapse
|
322
|
Development and physicochemical characterization of copper complexes-loaded PLGA nanoparticles. Int J Pharm 2009; 379:226-34. [PMID: 19428198 DOI: 10.1016/j.ijpharm.2009.03.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 03/17/2009] [Accepted: 03/27/2009] [Indexed: 11/24/2022]
Abstract
PLGA nanoparticles were prepared via a modified W/O/W emulsion solvent diffusion process, in which all formulation components were fully biocompatible and biodegradable. Different independent processing parameters were systematically studied. Nanoparticles were characterized by DLS (particle size, polydispersity, zeta-potential) and TEM/AFM (surface morphology). An optimized formulation was used to encapsulate copper complexes of cyclen and DOTA as potential PET imaging agents. Results showed that the predominant formulation factors appeared to be the lactide-to-glycolide (L:G) ratio of PLGA, the nature of the diffusion phase, and the presence of hydroxyl ions in the first-emulsion aqueous phase. By regulating those 3 parameters, PLGA nanoparticles were prepared with very good preparation yields (>95%), a size less than 200 nm and a polydispersity index less than 0.1. TEM pictures showed nanoparticles with a narrow size distribution, a spherical shape and a smooth surface. The optimized formulation allowed to encapsulate Cu-cyclen and Cu-DOTA complexes with an encapsulation efficiency between 20% and 25%.
Collapse
|
323
|
Park JH, von Maltzahn G, Zhang L, Derfus AM, Simberg D, Harris TJ, Ruoslahti E, Bhatia SN, Sailor MJ. Systematic surface engineering of magnetic nanoworms for in vivo tumor targeting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2009; 5:694-700. [PMID: 19263431 PMCID: PMC3058937 DOI: 10.1002/smll.200801789] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In the design of nanoparticles that can target disease tissue in vivo, parameters such as targeting ligand density, type of target receptor, and nanoparticle shape can play an important role in determining the extent of accumulation. Herein, a systematic study of these parameters for the targeting of mouse xenograft tumors is performed using superparamagnetic iron oxide as a model nanoparticle system. The type of targeting peptide (recognizing cell surface versus extracellular matrix), the surface coverage of the peptide, its attachment chemistry, and the shape of the nanomaterial [elongated (nanoworm, NW) versus spherical (nanosphere, NS)] are varied. Nanoparticle circulation times and in vivo tumor-targeting efficiencies are quantified in two xenograft models of human tumors (MDA-MB-435 human carcinoma and HT1080 human fibrosarcoma). It is found that the in vivo tumor-targeting ability of the NW is superior to that of the NS, that the smaller, neutral CREKA targeting group is more effective than the larger, positively charged F3 molecule, that a maximum in tumor-targeting efficiency and blood half-life is observed with approximately 60 CREKA peptides per NW for either the HT1080 or the MDA-MB-435 tumor types, and that incorporation of a 5-kDa polyethylene glycol linker improves targeting to both tumor types relative to a short linker. It is concluded that the blood half-life of a targeting molecule-nanomaterial ensemble is a key consideration when selecting the appropriate ligand and nanoparticle chemistry for tumor targeting.
Collapse
Affiliation(s)
- Ji-Ho Park
- Materials Science and Engineering Program, Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| | - Geoffrey von Maltzahn
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Lianglin Zhang
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Austin M. Derfus
- Department of Bioengineering, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| | - Dmitri Simberg
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Todd J. Harris
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Erkki Ruoslahti
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Sangeeta N. Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Michael J. Sailor
- Materials Science and Engineering Program, Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| |
Collapse
|
324
|
Walsh CL, Babin BM, Kasinskas RW, Foster JA, McGarry MJ, Forbes NS. A multipurpose microfluidic device designed to mimic microenvironment gradients and develop targeted cancer therapeutics. LAB ON A CHIP 2009; 9:545-54. [PMID: 19190790 PMCID: PMC2855303 DOI: 10.1039/b810571e] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The heterogeneity of cellular microenvironments in tumors severely limits the efficacy of most cancer therapies. We have designed a microfluidic device that mimics the microenvironment gradients present in tumors that will enable the development of more effective cancer therapies. Tumor cell masses were formed within micron-scale chambers exposed to medium perfusion on one side to create linear nutrient gradients. The optical accessibility of the PDMS and glass device enables quantitative transmitted and fluorescence microscopy of all regions of the cell masses. Time-lapse microscopy was used to measure the growth rate and show that the device can be used for long-term efficacy studies. Fluorescence microscopy was used to demonstrate that the cell mass contained viable, apoptotic, and acidic regions similar to in vivo tumors. The diffusion coefficient of doxorubicin was accurately measured, and the accumulation of therapeutic bacteria was quantified. The device is simple to construct, and it can easily be reproduced to create an array of in vitro tumors. Because microenvironment gradients and penetration play critical roles controlling drug efficacy, we believe that this microfluidic device will be vital for understanding the behavior of common cancer drugs in solid tumors and designing novel intratumorally targeted therapeutics.
Collapse
Affiliation(s)
- Colin L Walsh
- Department of Chemical Engineering, University of Massachusetts, 159 Goessmann Laboratory, 686 North Pleasant Street. Amherst, MA 01003-9303, USA
| | | | | | | | | | | |
Collapse
|
325
|
Targeting and imaging cancer cells by Folate-decorated, quantum dots (QDs)- loaded nanoparticles of biodegradable polymers. Biomaterials 2009; 30:1176-83. [DOI: 10.1016/j.biomaterials.2008.10.039] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Accepted: 10/23/2008] [Indexed: 01/10/2023]
|
326
|
Abstract
Cells within tissues are continuously exposed to physical forces including hydrostatic pressure, shear stress, and compression and tension forces. Cells dynamically adapt to force by modifying their behaviour and remodelling their microenvironment. They also sense these forces through mechanoreceptors and respond by exerting reciprocal actomyosin- and cytoskeletal-dependent cell-generated force by a process termed 'mechanoreciprocity'. Loss of mechanoreciprocity has been shown to promote the progression of disease, including cancer. Moreover, the mechanical properties of a tissue contribute to disease progression, compromise treatment and might also alter cancer risk. Thus, the changing force that cells experience needs to be considered when trying to understand the complex nature of tumorigenesis.
Collapse
Affiliation(s)
- Darci T Butcher
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
327
|
Nickel M, Strand SE, Lindén O, Wingårdh K, Tennwall J, Gleisner KS. Development and Evaluation of a Pharmacokinetic Model for Prediction of Radioimmunotherapy Based on Pretherapy Data. Cancer Biother Radiopharm 2009; 24:111-22. [DOI: 10.1089/cbr.2008.0530] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Mattias Nickel
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden
| | - Sven-Erik Strand
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden
| | - Ola Lindén
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Wingårdh
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden
| | - Jan Tennwall
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | | |
Collapse
|
328
|
Farias PMA, Santos BS, Fontes A. Semiconductor fluorescent quantum dots: efficient biolabels in cancer diagnostics. Methods Mol Biol 2009; 544:407-19. [PMID: 19488715 DOI: 10.1007/978-1-59745-483-4_27] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We present and discuss results and features related to the synthesis of water-soluble semiconductor quantum dots and their application as fluorescent biomarkers in cancer diagnostics. We have prepared and applied different core-shell quantum dots, such as cadmium telluride-cadmium sulfide, CdTe-CdS, and cadmium sulfide-cadmium hydroxide, CdS/Cd(OH)(2), in living healthy and neoplastic cells and tissues samples. The CdS/Cd(OH)(2) quantum dots presented the best results, maintaining high levels of luminescence as well as high photostability in cells and tissues. Labeled tissues and cells were analyzed by their resulting fluorescence, via conventional fluorescence microscopy or via laser scanning confocal microscopy. The procedure presented in this work was shown to be efficient as a potential tool for fast and precise cancer diagnostics.
Collapse
Affiliation(s)
- Patricia M A Farias
- Department of Biophysics and Radiobiology, Federal University of Pernambuco, Cidade Universitária, Recife, PE, Brazil.
| | | | | |
Collapse
|
329
|
Li ML, Wang JC, Schwartz JA, Gill-Sharp KL, Stoica G, Wang LV. In-vivo photoacoustic microscopy of nanoshell extravasation from solid tumor vasculature. JOURNAL OF BIOMEDICAL OPTICS 2009; 14:010507. [PMID: 19256687 PMCID: PMC6988903 DOI: 10.1117/1.3081556] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In this study, high resolution backward-mode photoacoustic microscopy (PAM) is used to noninvasively image progressive extravasation and accumulation of nanoshells within a solid tumor in vivo. PAM takes advantage of the strong near-infrared absorption of nanoshells and their extravasation tendency from leaky tumor vasculatures for imaging. Subcutaneous tumors are grown on immunocompetent BALB/c mice. Polyethylene glycol (PEGylated) nanoshells with a peak optical absorption at approximately 800 nm are intravenously administered. With an 800-nm laser source, a prescan prior to nanoshell injection is taken to determine the background that is free of nanoshell accumulation. After injection, the 3-D nanoshell distribution at the tumor foci is monitored by PAM for 6 h. Experimental results show that accumulated nanoshells delineate the tumor position. Nanoshell accumulation is heterogeneous in tumors: more concentrated within the tumor cortex and largely absent from the tumor core. Because nanoshells have been recently demonstrated to enhance thermal therapy of subcutaneous tumors, we anticipate that PAM will be an important aid before, during, and after nanoshell thermal therapy.
Collapse
Affiliation(s)
- Meng-Lin Li
- National Tsing Hua University, Department of Electrical Engineering, Hsinchu, 30013, Taiwan
| | | | | | | | - George Stoica
- Texas A&M University, Department of Veterinary Pathobiology, 5547 TAMU College Station, Texas 77843-5547
| | - Lihong V. Wang
- Washington University in St. Louis, Department of Biomedical Engineering, Optical Imaging Laboratory, St. Louis, Missouri 63130-4899
- Address all correspondence to: Lihong V. Wang, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130-4899. Tel: 314-935-6152; Fax: 314-935-7448;
| |
Collapse
|
330
|
Abstract
Shedding of cell surface antigens is an important biological process that is used by cells to modulate responses to signals in the extracellular environment. Because antibody-based therapies of cancer target cell surface antigens, it is important to understand more about the shedding process and how it affects tumor responses to this type of therapy. Up to now most attention has been focused on measuring the concentration of shed antigens in the blood and using these to determine the presence of a tumor and as a measure of response. The recent finding that the concentration of the tumor antigen mesothelin is extremely high within the interstitial space of tumors, where it can block antibody action, and that the concentration of shed mesothelin within the tumor is lowered by chemotherapy has important implications for the successful treatment of solid tumors by immunoconjugates and whole antibodies.
Collapse
Affiliation(s)
- Yujian Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-4264, USA
| | | |
Collapse
|
331
|
Nair A, Shen J, Thevenot P, Zou L, Cai T, Hu Z, Tang L. Enhanced intratumoral uptake of quantum dots concealed within hydrogel nanoparticles. NANOTECHNOLOGY 2008; 19:485102. [PMID: 21836292 DOI: 10.1088/0957-4484/19/48/485102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Effective nanomedical devices for tumor imaging and drug delivery are not yet available. In an attempt to construct a more functional device for tumor imaging, we have embedded quantum dots (which have poor circulatory behavior) within hydrogel nanoparticles made of poly-N-isopropylacrylamide. We found that the hydrogel encapsulated quantum dots are more readily taken up by cultured tumor cells. Furthermore, in a melanoma model, hydrogel encapsulated quantum dots also preferentially accumulate in the tumor tissue compared with normal tissue and have ∼16-fold greater intratumoral uptake compared to non-derivatized quantum dots. Our results suggest that these derivatized quantum dots, which have greatly improved tumor localization, may enhance cancer monitoring and chemotherapy.
Collapse
Affiliation(s)
- Ashwin Nair
- Joint Program in Bioengineering, University of Texas Southwestern Medical Center at Dallas and The University of Texas at Arlington, Arlington, TX 76019, USA
| | | | | | | | | | | | | |
Collapse
|
332
|
Patra CR, Cao S, Safgren S, Bhattacharya R, Ames MM, Shah V, Reid JM, Mukherjee P. Intracellular Fate of a Targeted Delivery System. J Biomed Nanotechnol 2008. [DOI: 10.1166/jbn.2008.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
333
|
Abstract
Nanoparticles have properties that are useful for the diagnosis and treatment of cancer, including their size-dependent properties, stability in solvent, ideal size for delivery within the body, and tunable surface chemistry for targeted delivery. Several different nanoparticle building blocks possessing varied functionality can be assembled into one multifunctional composite nanoparticle, further expanding their potential use in cancer diagnostics and therapeutics. Here, we present several examples of the types of functional composite nanoparticles that have been studied, in addition to highlighted applications of their uses.
Collapse
Affiliation(s)
- Pilar Rivera Gil
- Fachbereich Physik, Philipps Universitat Marburg, Marburg, Germany
| | | |
Collapse
|
334
|
|
335
|
Kang H, Alam R, Dixit V, Fisher M, Juliano RL. Cellular delivery and biological activity of antisense oligonucleotides conjugated to a targeted protein carrier. Bioconjug Chem 2008; 19:2182-8. [PMID: 18826264 PMCID: PMC2645933 DOI: 10.1021/bc800270w] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Targeted delivery can potentially improve the pharmacological effects of antisense and siRNA oligonucleotides. Here, we describe a novel bioconjugation approach to the delivery of splice-shifting antisense oligonucleotides (SSOs). The SSOs are linked to albumin via reversible S-S bonds. The albumin is also conjugated with poly(ethylene glycol) (PEG) chains that terminate in an RGD ligand that selectively binds the alphavbeta3 integrin. As a test system, we utilized human melanoma cells that express the alphavbeta3 integrin and that also contain a luciferase reporter gene that can be induced by delivery of SSOs to the cell nucleus. The RGD-PEG-SSO-albumin conjugates were endocytosed by the cells in an RGD-dependent manner; using confocal fluorescence microscopy, evidence was obtained that the SSOs accumulate in the nucleus. The conjugates were able to robustly induce luciferase expression at concentrations in the 25-200 nM range. At these levels, little short-term or long-term toxicity was observed. Thus, the RGD-PEG-albumin conjugates may provide an effective tool for targeted delivery of oligonucleotides to certain cells and tissues.
Collapse
Affiliation(s)
- Hyunmin Kang
- Division of Molecular Pharmaceutics School of Pharmacy CB 7360 University of North Carolina Chapel Hill NC 27599
| | - Rowshon Alam
- Division of Molecular Pharmaceutics School of Pharmacy CB 7360 University of North Carolina Chapel Hill NC 27599
| | - Vidula Dixit
- Division of Molecular Pharmaceutics School of Pharmacy CB 7360 University of North Carolina Chapel Hill NC 27599
| | - Michael Fisher
- Division of Molecular Pharmaceutics School of Pharmacy CB 7360 University of North Carolina Chapel Hill NC 27599
| | - Rudy L Juliano
- Division of Molecular Pharmaceutics School of Pharmacy CB 7360 University of North Carolina Chapel Hill NC 27599
| |
Collapse
|
336
|
Manocha B, Margaritis A. Production and Characterization of γ-Polyglutamic Acid Nanoparticles for Controlled Anticancer Drug Release. Crit Rev Biotechnol 2008; 28:83-99. [DOI: 10.1080/07388550802107483] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
337
|
Sharma G, Anabousi S, Ehrhardt C, Ravi Kumar MNV. Liposomes as targeted drug delivery systems in the treatment of breast cancer. J Drug Target 2008; 14:301-10. [PMID: 16882550 DOI: 10.1080/10611860600809112] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Solid tumors such as breast cancer have historically provided many challenges to anti-cancer therapy. Therapeutic hurdles to drug penetration in solid tumors include heterogeneous vascular supply and high interstitial pressures within tumor tissue, particularly in necrotic zones, lower pH and presence of leaky vasculature leading to reduced therapeutic response. Liposome based drug delivery systems offer the potential to enhance the therapeutic index of anti-cancer agents, either by increasing the drug concentration in tumor cells and/or by decreasing the exposure in normal tissues exploiting enhanced permeability and retention effect phenomenon and by utilizing targeting strategies. This review discusses recent trends in liposome-based drug delivery system both for diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- G Sharma
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S. A. S. Nagar, 160 062 Punjab, India
| | | | | | | |
Collapse
|
338
|
Goodman TT, Ng CP, Pun SH. 3-D tissue culture systems for the evaluation and optimization of nanoparticle-based drug carriers. Bioconjug Chem 2008; 19:1951-9. [PMID: 18788773 PMCID: PMC2652657 DOI: 10.1021/bc800233a] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nanoparticle carriers are attractive vehicles for a variety of drug delivery applications. In order to evaluate nanoparticle formulations for biological efficacy, monolayer cell cultures are typically used as in vitro testing platforms. However, these studies sometimes poorly predict the efficacy of the drug in vivo. The poor in vitro and in vivo correlation may be attributed in part to the inability of two-dimensional cultures to reproduce extracellular barriers, and may also be due to differences in cell phenotype between cells cultured as monolayers and cells in native tissue. In order to more accurately predict in vivo results, it is desirable to test nanoparticle therapeutics in cells cultured in three-dimensional (3-D) models that mimic in vivo conditions. In this review, we discuss some 3-D culture systems that have been used to assess nanoparticle delivery and highlight several implications for nanoparticle design garnered from studies using these systems. While our focus will be on nanoparticle drug formulations, many of the systems discussed here could, or have been, used for the assessment of small molecule or peptide/protein drugs. We also offer some examples of advancements in 3-D culture that could provide even more highly predictive data for designing nanoparticle therapeutics for in vivo applications.
Collapse
Affiliation(s)
| | | | - Suzie Hwang Pun
- Department of Bioengineering, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| |
Collapse
|
339
|
Karathanasis E, Chan L, Balusu SR, D'Orsi CJ, Annapragada AV, Sechopoulos I, Bellamkonda RV. Multifunctional nanocarriers for mammographic quantification of tumor dosing and prognosis of breast cancer therapy. Biomaterials 2008; 29:4815-22. [PMID: 18814908 DOI: 10.1016/j.biomaterials.2008.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 08/22/2008] [Indexed: 10/21/2022]
Abstract
Nanoscale therapeutic interventions are increasingly important elements in the portfolio of cancer therapeutics. The efficacy of nanotherapeutics is dictated, in part, by the access they have to tumors via the leaky tumor vasculature. Yet, the extent of tumor vessel leakiness in individual tumors varies widely resulting in a correspondingly wide tumor dosing and resulting range of responses to therapy. Here we report the design of a multifunctional nanocarrier that simultaneously encapsulates a chemotherapeutic and a contrast agent which enables a personalized nanotherapeutic approach for breast cancer therapy by permitting tracking of the nanocarrier distribution by mammography, a widely used imaging modality. Following systemic administration in a rat breast tumor model, imaging demonstrated a wide range of intratumoral deposition of the nanocarriers, indicating variable tumor vessel leakiness. Notably, specific tumors that exhibited high uptake of the nanocarrier as visualized by imaging were precisely the animals that responded best to the treatment as quantified by low tumor growth and prolonged survival.
Collapse
Affiliation(s)
- Efstathios Karathanasis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | | | | | | | | | | | | |
Collapse
|
340
|
Park JH, von Maltzahn G, Ruoslahti E, Bhatia S, Sailor M. Micellar Hybrid Nanoparticles for Simultaneous Magnetofluorescent Imaging and Drug Delivery. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200801810] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
341
|
Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, Babb JS, Schneider RJ, Formenti SC, Dustin ML, Demaria S. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:3099-107. [PMID: 18713980 PMCID: PMC2587101 DOI: 10.4049/jimmunol.181.5.3099] [Citation(s) in RCA: 556] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recruitment of effector T cells to inflamed peripheral tissues is regulated by chemokines and their receptors, but the factors regulating recruitment to tumors remain largely undefined. Ionizing radiation (IR) therapy is a common treatment modality for breast and other cancers. Used as a cytocidal agent for proliferating cancer cells, IR in combination with immunotherapy has been shown to promote immune-mediated tumor destruction in preclinical studies. In this study we demonstrate that IR markedly enhanced the secretion by mouse and human breast cancer cells of CXCL16, a chemokine that binds to CXCR6 on Th1 and activated CD8 effector T cells, and plays an important role in their recruitment to sites of inflammation. Using a poorly immunogenic mouse model of breast cancer, we found that irradiation increased the migration of CD8(+)CXCR6(+) activated T cells to tumors in vitro and in vivo. CXCR6-deficient mice showed reduced infiltration of tumors by activated CD8 T cells and impaired tumor regression following treatment with local IR to the tumor and Abs blocking the negative regulator of T cell activation, CTLA-4. These results provide the first evidence that IR can induce the secretion by cancer cells of proinflammatory chemotactic factors that recruit antitumor effector T cells. The ability of IR to convert tumors into "inflamed" peripheral tissues could be exploited to overcome obstacles at the effector phase of the antitumor immune response and improve the therapeutic efficacy of immunotherapy.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- CD8-Positive T-Lymphocytes/physiology
- CD8-Positive T-Lymphocytes/radiation effects
- Cell Line, Tumor
- Chemokine CXCL16
- Chemokine CXCL6/metabolism
- Chemokine CXCL6/radiation effects
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Chemokines, CXC/radiation effects
- Chemotaxis, Leukocyte/drug effects
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Radiation, Ionizing
- Receptors, CXCR
- Receptors, CXCR6
- Receptors, Scavenger/genetics
- Receptors, Scavenger/radiation effects
- Up-Regulation/radiation effects
Collapse
Affiliation(s)
- Satoko Matsumura
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Baomei Wang
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Noriko Kawashima
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Steve Braunstein
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Michelle Badura
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Thomas O. Cameron
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, and the Program in Molecular Pathogenesis, The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, 540 First Avenue, NY 10016
| | - James S. Babb
- Department of Radiology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Robert J. Schneider
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Silvia C. Formenti
- Department of Radiation Oncology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| | - Michael L. Dustin
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, and the Program in Molecular Pathogenesis, The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, 540 First Avenue, NY 10016
| | - Sandra Demaria
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016
| |
Collapse
|
342
|
Thurber GM, Schmidt MM, Wittrup KD. Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance. Adv Drug Deliv Rev 2008; 60:1421-34. [PMID: 18541331 PMCID: PMC2820307 DOI: 10.1016/j.addr.2008.04.012] [Citation(s) in RCA: 436] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Accepted: 04/16/2008] [Indexed: 01/01/2023]
Abstract
Antibodies have proven to be effective agents in cancer imaging and therapy. One of the major challenges still facing the field is the heterogeneous distribution of these agents in tumors when administered systemically. Large regions of untargeted cells can therefore escape therapy and potentially select for more resistant cells. We present here a summary of theoretical and experimental approaches to analyze and improve antibody penetration in tumor tissue.
Collapse
Affiliation(s)
- Greg M Thurber
- Department Chemical Engineering, Massachusetts Institute of Technology, USA
| | | | | |
Collapse
|
343
|
Decuzzi P, Pasqualini R, Arap W, Ferrari M. Intravascular delivery of particulate systems: does geometry really matter? Pharm Res 2008; 26:235-43. [PMID: 18712584 DOI: 10.1007/s11095-008-9697-x] [Citation(s) in RCA: 411] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Accepted: 07/24/2008] [Indexed: 10/21/2022]
Abstract
In cancer therapy and imaging, the systemic passive delivery of particulate systems has relied on the enhanced permeability and retention (EPR) effect: sufficiently small particles can cross the endothelial fenestrations and accumulate in the tumor parenchyma. The vast majority of man-made particulates exhibit a spherical shape as a result of surface energy minimization during their synthesis. The advent of phage display libraries, which are revealing the extraordinary molecular diversity of endothelial cells, and the development of processes for fabricating particles with shapes other than spherical are opening the path to new design solutions for systemically administered targeted particulates. In this paper, the role of particle geometry (i.e., size and shape) is discussed at the tissue and cellular scales. Emphasis is placed on how the synergistic effect of particle geometry and molecular targeting can enhance the specificity of delivery. The intravascular delivery process has been broken into three events: margination, firm adhesion and control of internalization. Predictions from mathematical models and observations from in-vitro experiments were used to show the relevance of particle geometry in systemic delivery. Rational design of particulate systems should consider, beside the physico-chemical properties of the surface coatings, geometrical features as size and shape. The integration of mathematical modeling with in-vitro and in-vivo testing provides the tools for establishing a rational design of nanoparticles.
Collapse
Affiliation(s)
- Paolo Decuzzi
- School of Health Information Sciences, The University of Texas Health Science Center, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|
344
|
Smith AM, Duan H, Mohs AM, Nie S. Bioconjugated quantum dots for in vivo molecular and cellular imaging. Adv Drug Deliv Rev 2008; 60:1226-1240. [PMID: 18495291 PMCID: PMC2649798 DOI: 10.1016/j.addr.2008.03.015] [Citation(s) in RCA: 743] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 03/12/2008] [Indexed: 01/08/2023]
Abstract
Semiconductor quantum dots (QDs) are tiny light-emitting particles on the nanometer scale, and are emerging as a new class of fluorescent labels for biology and medicine. In comparison with organic dyes and fluorescent proteins, they have unique optical and electronic properties, with size-tunable light emission, superior signal brightness, resistance to photobleaching, and broad absorption spectra for simultaneous excitation of multiple fluorescence colors. QDs also provide a versatile nanoscale scaffold for designing multifunctional nanoparticles with both imaging and therapeutic functions. When linked with targeting ligands such as antibodies, peptides or small molecules, QDs can be used to target tumor biomarkers as well as tumor vasculatures with high affinity and specificity. Here we discuss the synthesis and development of state-of-the-art QD probes and their use for molecular and cellular imaging. We also examine key issues for in vivo imaging and therapy, such as nanoparticle biodistribution, pharmacokinetics, and toxicology.
Collapse
Affiliation(s)
- Andrew M. Smith
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle, Suite 2001, Atlanta, GA 30322, USA
| | - Hongwei Duan
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle, Suite 2001, Atlanta, GA 30322, USA
| | - Aaron M. Mohs
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle, Suite 2001, Atlanta, GA 30322, USA
| | - Shuming Nie
- Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, 101 Woodruff Circle, Suite 2001, Atlanta, GA 30322, USA
| |
Collapse
|
345
|
Karathanasis E, Park J, Agarwal A, Patel V, Zhao F, Annapragada AV, Hu X, Bellamkonda RV. MRI mediated, non-invasive tracking of intratumoral distribution of nanocarriers in rat glioma. NANOTECHNOLOGY 2008; 19:315101. [PMID: 21828778 DOI: 10.1088/0957-4484/19/31/315101] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Nanocarrier mediated therapy of gliomas has shown promise. The success of systemic nanocarrier-based chemotherapy is critically dependent on the so-called leaky vasculature to permit drug extravasation across the blood-brain barrier. Yet, the extent of vascular permeability in individual tumors varies widely, resulting in a correspondingly wide range of responses to the therapy. However, there exist no tools currently for rationally determining whether tumor blood vessels are amenable to nanocarrier mediated therapy in an individualized, patient specific manner today. To address this need for brain tumor therapy, we have developed a multifunctional 100 nm scale liposomal agent encapsulating a gadolinium-based contrast agent for contrast-enhanced magnetic resonance imaging with prolonged blood circulation. Using a 9.4 T MRI system, we were able to track the intratumoral distribution of the gadolinium-loaded nanocarrier in a rat glioma model for a period of three days due to improved magnetic properties of the contrast agent being packaged in a nanocarrier. Such a nanocarrier provides a tool for non-invasively assessing the suitability of tumors for nanocarrier mediated therapy and then optimizing the treatment protocol for each individual tumor. Additionally, the ability to image the tumor in high resolution can potentially constitute a surgical planning tool for tumor resection.
Collapse
Affiliation(s)
- Efstathios Karathanasis
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | | | | | | | | | | | | | | |
Collapse
|
346
|
Bawarski WE, Chidlowsky E, Bharali DJ, Mousa SA. Emerging nanopharmaceuticals. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2008; 4:273-82. [PMID: 18640076 DOI: 10.1016/j.nano.2008.06.002] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 05/13/2008] [Accepted: 06/03/2008] [Indexed: 12/31/2022]
Abstract
A budding interest in nanopharmaceuticals has generated a number of advancements throughout recent years with a focus on engineering novel applications. Nanotechnology also offers the ability to detect diseases at much earlier stages, such as finding hidden or overt metastatic colonies often seen in patients diagnosed with breast, lung, colon, prostate, and ovarian cancer. Diagnostic applications could build upon conventional procedures using nanoparticles, such as colloidal gold, iron oxide crystals, and quantum dots. Additionally, diseases may be managed by multifunctional agents encompassing both imaging and therapeutic capabilities, thus allowing simultaneous monitoring and treatment. A detailed evaluation of each formulation is essential to expand our current nanopharmaceutical repertoire. However, the safety and long-term effects of nanoformulations must not be overlooked. This review will provide a brief discussion of the major nanopharmaceutical formulations as well as the impact of nanotechnology into the future.
Collapse
Affiliation(s)
- Willie E Bawarski
- Pharmaceutical Research Institute at Albany College of Pharmacy, Rensselaer, New York, USA
| | | | | | | |
Collapse
|
347
|
Venkatasubramanian R, Henson MA, Forbes NS. Integrating cell-cycle progression, drug penetration and energy metabolism to identify improved cancer therapeutic strategies. J Theor Biol 2008; 253:98-117. [PMID: 18402980 PMCID: PMC2517579 DOI: 10.1016/j.jtbi.2008.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 02/12/2008] [Accepted: 02/12/2008] [Indexed: 12/26/2022]
Abstract
The effectiveness of chemotherapeutic drugs in tumors is reduced by multiple effects including drug diffusion and variable susceptibility of local cell populations. We hypothesized that quantifying the interactions between drugs and tumor microenvironments could be used to identify more effective anti-cancer strategies. To test this hypothesis we created a mathematical model that integrated intracellular metabolism, nutrient and drug diffusion, cell-cycle progression, cellular drug effects, and drug pharmacokinetics. To our knowledge, this is the first model that combines these elements and has coupled them to experimentally derived parameters. Drug cytotoxicity was assumed to be cell-cycle phase specific, and progression through the cell cycle was assumed to be dependent on ATP generation. The model consisted of a coupled set of nonlinear partial differential, ordinary differential and algebraic equations with an outer free boundary, which was solved using orthogonal collocation on a moving grid of finite elements. Model simulations showed the existence of an optimum drug diffusion coefficient: a low diffusivity prevents effective penetration before the drug is cleared from the blood and a high diffusivity limits drug retention. This result suggests that increasing the molecular weight of the anti-cancer drug paclitaxel from 854 to approximately 20,000 by nanoparticle conjugation would improve its efficacy. The simulations also showed that fast growing tumors are less responsive to therapy than are slower tumors with more quiescent cells, demonstrating the competing effects of regrowth and cytotoxicity. The therapeutic implications of the simulation results are that (1) monolayer cultures are inadequate for accurately determining therapeutic effects in vitro, (2) decreasing the diffusivity of paclitaxel could increase its efficacy, and (3) measuring the proliferation fraction in tumors could enhance the prediction of therapeutic efficacy.
Collapse
Affiliation(s)
- Raja Venkatasubramanian
- Department of Chemical Engineering, University of Massachusetts, 159 Goessmann Hall, Amherst, 686 North Pleasant Street, Amherst, MA 01003-9303, USA
| | | | | |
Collapse
|
348
|
Ackerman ME, Pawlowski D, Wittrup KD. Effect of antigen turnover rate and expression level on antibody penetration into tumor spheroids. Mol Cancer Ther 2008; 7:2233-40. [PMID: 18645032 PMCID: PMC2831054 DOI: 10.1158/1535-7163.mct-08-0067] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Poor tissue penetration is a significant obstacle to the development of successful antibody drugs for immunotherapy of solid tumors, and diverse alterations to the properties of antibody drugs have been made to improve penetration and homogeneity of exposure. However, in addition to properties of the antibody drug, mathematical models of antibody transport predict that the antigen expression level and turnover rate significantly influence penetration. As intrinsic antigen properties are likely to be difficult to modify, they may set inherent limits to penetration. Accordingly, in this study, we assess their contribution by evaluating the distance to which antibodies penetrate spheroids when these antigen properties are systematically varied. Additionally, the penetration profiles of antibodies against carcinoembryonic antigen and A33, two targets of clinical interest, are compared. The results agree well with the quantitative predictions of the model and show that localizing antibody to distal regions of tumors is best achieved by selecting slowly internalized targets that are not expressed above the level necessary for recruiting a toxic dose of therapeutic. Each antibody-bound antigen molecule that is turned over or present in excess incurs a real cost in terms of penetration depth-a limiting factor in the development of effective therapies for treating solid tumors.
Collapse
Affiliation(s)
- Margaret E. Ackerman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - David Pawlowski
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, Ohio
| | - K. Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
349
|
Chang MY, Shiau AL, Chen YH, Chang CJ, Chen HHW, Wu CL. Increased apoptotic potential and dose-enhancing effect of gold nanoparticles in combination with single-dose clinical electron beams on tumor-bearing mice. Cancer Sci 2008; 99:1479-84. [PMID: 18410403 PMCID: PMC11158140 DOI: 10.1111/j.1349-7006.2008.00827.x] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 02/27/2008] [Accepted: 03/10/2008] [Indexed: 11/27/2022] Open
Abstract
High atomic number material, such as gold, may be used in conjunction with radiation to provide dose enhancement in tumors. In the current study, we investigated the dose-enhancing effect and apoptotic potential of gold nanoparticles in combination with single-dose clinical electron beams on B16F10 melanoma tumor-bearing mice. We revealed that the accumulation of gold nanoparticles was detected inside B16F10 culture cells after 18 h of incubation, and moreover, the gold nanoparticles were shown to be colocalized with endoplasmic reticulum and Golgi apparatus in cells. Furthermore, gold nanoparticles radiosensitized melanoma cells in the colony formation assay (P = 0.02). Using a B16F10 tumor-bearing mouse model, we further demonstrated that gold nanoparticles in conjunction with ionizing radiation significantly retarded tumor growth and prolonged survival compared to the radiation alone controls (P < 0.05). Importantly, an increase of apoptotic signals was detected inside tumors in the combined treatment group (P < 0.05). Knowing that radiation-induced apoptosis has been considered a determinant of tumor responses to radiation therapy, and the length of tumor regrowth delay correlated with the extent of apoptosis after single-dose radiotherapy, these results may suggest the clinical potential of gold nanoparticles in improving the outcome of melanoma radiotherapy.
Collapse
Affiliation(s)
- Meng-Ya Chang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University Medical College, 1 Dashiue Road, Tainan 701, Taiwan
| | | | | | | | | | | |
Collapse
|
350
|
Diagaradjane P, Orenstein-Cardona JM, Colón-Casasnovas NE, Deorukhkar A, Shentu S, Kuno N, Schwartz DL, Gelovani JG, Krishnan S. Imaging epidermal growth factor receptor expression in vivo: pharmacokinetic and biodistribution characterization of a bioconjugated quantum dot nanoprobe. Clin Cancer Res 2008; 14:731-41. [PMID: 18245533 DOI: 10.1158/1078-0432.ccr-07-1958] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To develop and validate an optical imaging nanoprobe for the discrimination of epidermal growth factor (EGF) receptor (EGFR)-overexpressing tumors from surrounding normal tissues that also expresses EGFR. EXPERIMENTAL DESIGN Near-infrared (NIR) quantum dots (QD) were coupled to EGF using thiol-maleimide conjugation to create EGF-QD nanoprobes. In vitro binding affinity of these nanoprobes and unconjugated QDs was evaluated in a panel of cell lines, with and without anti-EGFR antibody pretreatment. Serial optical imaging of HCT116 xenograft tumors was done after systemic injection of QD and EGF-QD. RESULTS EGF-QD showed EGFR-specific binding in vitro. In vivo imaging showed three distinct phases, tumor influx ( approximately 3 min), clearance ( approximately 60 min), and accumulation (1-6 h), of EGF-QD nanoprobes. Both QD and EGF-QD showed comparable nonspecific rapid tumor influx and clearance followed by attainment of an apparent dynamic equilibrium at approximately 60 min. Subsequently (1-6 h), whereas QD concentration gradually decreased in tumors, EGF-QDs progressively accumulated in tumors. On delayed imaging at 24 h, tumor fluorescence decreased to near-baseline levels for both QD and EGF-QD. Ex vivo whole-organ fluorescence, tissue homogenate fluorescence, and confocal microscopic analyses confirmed tumor-specific accumulation of EGF-QD at 4 h. Immunofluorescence images showed diffuse colocalization of EGF-QD fluorescence within EGFR-expressing tumor parenchyma compared with patchy perivascular sequestration of QD. CONCLUSION These results represent the first pharmacokinetic characterization of a robust EGFR imaging nanoprobe. The measurable contrast enhancement of tumors 4 h after systemic administration of EGF-QD and its subsequent normalization at 24 h imply that this nanoprobe may permit quantifiable and repetitive imaging of EGFR expression.
Collapse
Affiliation(s)
- Parmeswaran Diagaradjane
- Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|