151
|
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|
152
|
Solarska-Ściuk K, Gajewska A, Glińska S, Michlewska S, Balcerzak Ł, Jamrozik A, Skolimowski J, Burda K, Bartosz G. Effect of functionalized and non-functionalized nanodiamond on the morphology and activities of antioxidant enzymes of lung epithelial cells (A549). Chem Biol Interact 2014; 222:135-47. [DOI: 10.1016/j.cbi.2014.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 09/02/2014] [Accepted: 10/06/2014] [Indexed: 11/27/2022]
|
153
|
Toy R, Bauer L, Hoimes C, Ghaghada KB, Karathanasis E. Targeted nanotechnology for cancer imaging. Adv Drug Deliv Rev 2014; 76:79-97. [PMID: 25116445 PMCID: PMC4169743 DOI: 10.1016/j.addr.2014.08.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/26/2014] [Accepted: 08/04/2014] [Indexed: 02/02/2023]
Abstract
Targeted nanoparticle imaging agents provide many benefits and new opportunities to facilitate accurate diagnosis of cancer and significantly impact patient outcome. Due to the highly engineerable nature of nanotechnology, targeted nanoparticles exhibit significant advantages including increased contrast sensitivity, binding avidity and targeting specificity. Considering the various nanoparticle designs and their adjustable ability to target a specific site and generate detectable signals, nanoparticles can be optimally designed in terms of biophysical interactions (i.e., intravascular and interstitial transport) and biochemical interactions (i.e., targeting avidity towards cancer-related biomarkers) for site-specific detection of very distinct microenvironments. This review seeks to illustrate that the design of a nanoparticle dictates its in vivo journey and targeting of hard-to-reach cancer sites, facilitating early and accurate diagnosis and interrogation of the most aggressive forms of cancer. We will report various targeted nanoparticles for cancer imaging using X-ray computed tomography, ultrasound, magnetic resonance imaging, nuclear imaging and optical imaging. Finally, to realize the full potential of targeted nanotechnology for cancer imaging, we will describe the challenges and opportunities for the clinical translation and widespread adaptation of targeted nanoparticles imaging agents.
Collapse
Affiliation(s)
- Randall Toy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lisa Bauer
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Physics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Christopher Hoimes
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Ketan B Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX 77030, USA; Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
154
|
Ozsun O, Thompson RL, Ekinci KL, Tien J. Non-invasive mapping of interstitial fluid pressure in microscale tissues. Integr Biol (Camb) 2014; 6:979-87. [PMID: 25181983 DOI: 10.1039/c4ib00164h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study describes a non-invasive method for mapping interstitial fluid pressure within hydrogel-based microscale tissues. The method is based on embedding (or forming) a tissue within a silicone (PDMS) microfluidic device, and measuring the extremely slight displacement (<1 μm) of the PDMS optically when the device is pressurized under static and flow conditions. The displacement field under uniform pressure provides a map of the local device stiffness, which can then be used to obtain the non-uniform pressure field under flow conditions. We have validated this method numerically and applied it towards determining the hydraulic properties of tumor cell aggregates, blind-ended epithelial tubes, and perfused endothelial tubes that were all cultured within micropatterned collagen gels. The method provides an accessible tool for generating high-resolution maps of interstitial fluid pressure for studies in mechanobiology.
Collapse
Affiliation(s)
- Ozgur Ozsun
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | | | | | | |
Collapse
|
155
|
Kwak B, Ozcelikkale A, Shin CS, Park K, Han B. Simulation of complex transport of nanoparticles around a tumor using tumor-microenvironment-on-chip. J Control Release 2014; 194:157-67. [PMID: 25194778 DOI: 10.1016/j.jconrel.2014.08.027] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 08/14/2014] [Accepted: 08/26/2014] [Indexed: 12/28/2022]
Abstract
Delivery of therapeutic agents selectively to tumor tissue, which is referred as "targeted delivery," is one of the most ardently pursued goals of cancer therapy. Recent advances in nanotechnology enable numerous types of nanoparticles (NPs) whose properties can be designed for targeted delivery to tumors. In spite of promising early results, the delivery and therapeutic efficacy of the majority of NPs are still quite limited. This is mainly attributed to the limitation of currently available tumor models to test these NPs and systematically study the effects of complex transport and pathophysiological barriers around the tumors. In this study, thus, we developed a new in vitro tumor model to recapitulate the tumor microenvironment determining the transport around tumors. This model, named tumor-microenvironment-on-chip (T-MOC), consists of 3-dimensional microfluidic channels where tumor cells and endothelial cells are cultured within extracellular matrix under perfusion of interstitial fluid. Using this T-MOC platform, the transport of NPs and its variation due to tumor microenvironmental parameters have been studied including cut-off pore size, interstitial fluid pressure, and tumor tissue microstructure. The results suggest that T-MOC is capable of simulating the complex transport around the tumor, and providing detailed information about NP transport behavior. This finding confirms that NPs should be designed considering their dynamic interactions with tumor microenvironment.
Collapse
Affiliation(s)
- Bongseop Kwak
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Altug Ozcelikkale
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Crystal S Shin
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Kinam Park
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
156
|
Magnetic Fe3O4 nanoparticles grafted with single-chain antibody (scFv) and docetaxel loaded β-cyclodextrin potential for ovarian cancer dual-targeting therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 42:325-32. [DOI: 10.1016/j.msec.2014.05.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/13/2014] [Accepted: 05/18/2014] [Indexed: 11/21/2022]
|
157
|
Budker VG, Monahan SD, Subbotin VM. Loco-regional cancer drug therapy: present approaches and rapidly reversible hydrophobization (RRH) of therapeutic agents as the future direction. Drug Discov Today 2014; 19:1855-70. [PMID: 25173702 DOI: 10.1016/j.drudis.2014.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023]
Abstract
Insufficient drug uptake by solid tumors remains the major problem for systemic chemotherapy. Many studies have demonstrated anticancer drug effects to be dose-dependent, although dose-escalation studies have resulted in limited survival benefit with increased systemic toxicities. One solution to this has been the idea of loco-regional drug treatments, which offer dramatically higher drug concentrations in tumor tissues while minimizing systemic toxicity. Although loco-regional delivery has been most prominent in cancers of the liver, soft tissues and serosal peritoneal malignancies, survival benefits are very far from desirable. This review discusses the evolution of loco-regional treatments, the present approaches and offers rapidly reversible hydrophobization of drugs as the new future direction.
Collapse
|
158
|
Bhatnagar S, Deschenes E, Liao J, Cilliers C, Thurber GM. Multichannel imaging to quantify four classes of pharmacokinetic distribution in tumors. J Pharm Sci 2014; 103:3276-86. [PMID: 25048378 DOI: 10.1002/jps.24086] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/12/2014] [Accepted: 06/16/2014] [Indexed: 01/31/2023]
Abstract
Low and heterogeneous delivery of drugs and imaging agents to tumors results in decreased efficacy and poor imaging results. Systemic delivery involves a complex interplay of drug properties and physiological factors, and heterogeneity in the tumor microenvironment makes predicting and overcoming these limitations exceptionally difficult. Theoretical models have indicated that there are four different classes of pharmacokinetic behavior in tissue, depending on the fundamental steps in distribution. In order to study these limiting behaviors, we used multichannel fluorescence microscopy and stitching of high-resolution images to examine the distribution of four agents in the same tumor microenvironment. A validated generic partial differential equation model with a graphical user interface was used to select fluorescent agents exhibiting these four classes of behavior, and the imaging results agreed with predictions. BODIPY-FL exhibited higher concentrations in tissue with high blood flow, cetuximab gave perivascular distribution limited by permeability, high plasma protein and target binding resulted in diffusion-limited distribution for Hoechst 33342, and Integrisense 680 was limited by the number of binding sites in the tissue. Together, the probes and simulations can be used to investigate distribution in other tumor models, predict tumor drug distribution profiles, and design and interpret in vivo experiments.
Collapse
Affiliation(s)
- Sumit Bhatnagar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| | | | | | | | | |
Collapse
|
159
|
Nguyen DT, Kim KS. Functionalization of magnetic nanoparticles for biomedical applications. KOREAN J CHEM ENG 2014. [DOI: 10.1007/s11814-014-0156-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
160
|
Redolfi Riva E, Desii A, Sinibaldi E, Ciofani G, Piazza V, Mazzolai B, Mattoli V. Gold nanoshell/polysaccharide nanofilm for controlled laser-assisted tissue thermal ablation. ACS NANO 2014; 8:5552-5563. [PMID: 24797875 DOI: 10.1021/nn406348v] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
We report on the fabrication and characterization of a freestanding ultrathin, mucoadhesive gold nanoshell/polysaccharide multilayer nanocomposite (thermonanofilm, TNF), that can be used for controlled photothermal ablation of tissues through irradiation with near-infrared radiation (NIR) laser. The aim of this work is to provide a new strategy to precisely control particle concentration during photothermalization of cancerous lesions, since unpredictable and aspecific biodistributions still remains the central issue of inorganic nanoparticle-assisted photothermal ablation. Gold nanoshell encapsulation in polysaccharide matrix is achieved by drop casting deposition method combined with spin-assisted layer-by-layer (LbL) assembly. Submicrometric thickness of films ensures tissue adhesion. Basic laser-induced heating functionality has been demonstrated by in vitro TNF-mediated thermal ablation of human neuroblastoma cancer cells, evidenced by irreversible damage to cell membranes and nuclei. Ex vivo localized vaporization and carbonization of animal muscular tissue is also demonstrated by applying TNF onto tissue surface. Thermal distribution in the tissue reaches a steady state in a few seconds, with significant increases in temperature (ΔT > 50) occurring across an 1 mm span, ensuring control of local photothermalization and providing more safety and predictability with respect to traditional laser surgery. A steady-state model of tissue thermalization mediated by TNFs is also introduced, predicting the temperature distribution being known the absorbance of TNFs, the laser power, and the tissue thermal conductivity, thus providing useful guidelines in the development of TNFs. Thermonanofilms can find applications for local photothermal treatment of cancerous lesions and wherever high precision and control of heat treatment is required.
Collapse
Affiliation(s)
- Eugenio Redolfi Riva
- Center for Micro-BioRobotics, Istituto Italiano di Tecnologia , Pontedera, 56025, Italy
| | | | | | | | | | | | | |
Collapse
|
161
|
Hollevoet K, Mason-Osann E, Liu XF, Imhof-Jung S, Niederfellner G, Pastan I. In vitro and in vivo activity of the low-immunogenic antimesothelin immunotoxin RG7787 in pancreatic cancer. Mol Cancer Ther 2014; 13:2040-9. [PMID: 24928849 DOI: 10.1158/1535-7163.mct-14-0089-t] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, and new therapies are needed. RG7787 is a novel low-immunogenic antimesothelin recombinant immunotoxin (RIT), engineered to overcome the limitations of SS1P, a RIT now in clinical trials. In vitro activity was evaluated on five established PDAC cell lines (KLM-1, AsPC-1, BxPC-3, Panc 3.014, and PK-1) and on PDAC cells directly established from a patient tumor (GUMC108). RG7787 had subnanomolar IC50s in most cell lines, and was significantly more active than SS1P in GUMC108, KLM-1, and Panc 3.014 cells. GUMC108 was most sensitive, with RG7787 killing >99% of the cells. In a subcutaneous KLM-1 xenograft mouse model, two cycles of 3 × 2.5 mg/kg RG7787 QOD combined with two cycles of 1 × 50 mg/kg paclitaxel induced near-complete responses, with all tumors regressing below 5 mm(3) within 30 days after therapy was initiated (>95% decrease) and no significant growth increase for at least another 3 weeks. RG7787 alone gave limited but significant regressions and paclitaxel by itself arrested tumor growth. Quantifying the uptake of Alexa Fluor 647-labeled RG7787 in tumors showed that the RIT reached only 45% of KLM-1 cells, accounting in part for the limited responses. Paclitaxel did not improve RG7787 uptake, which thus cannot explain the beneficial effect of the combination therapy. In conclusion, RG7787 has high cytotoxic activity on PDAC cell lines as well as on primary patient cells. In vivo, this novel RIT gives durable near-complete tumor responses when combined with paclitaxel. RG7787 merits further evaluation for the treatment of PDAC.
Collapse
Affiliation(s)
- Kevin Hollevoet
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland; Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; and
| | - Emily Mason-Osann
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Xiu-fen Liu
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sabine Imhof-Jung
- Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Penzberg, Germany
| | - Gerhard Niederfellner
- Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Penzberg, Germany
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland;
| |
Collapse
|
162
|
Intracellular transport of nanodiamond particles in human endothelial and epithelial cells. Chem Biol Interact 2014; 219:90-100. [PMID: 24882084 DOI: 10.1016/j.cbi.2014.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 04/13/2014] [Accepted: 05/20/2014] [Indexed: 11/20/2022]
Abstract
During the recent years nanodiamonds have been the subject of interest as possible means of targeted delivery of anticancer substances. Detonation nanodiamonds are attractive candidates for intracellular studies due to their synthesis methods, low cost, good biocompatibility and facile surface functionalizability. Our previous study, in which we used nanoparticles obtained by different methods showed the significance of size and way of production of nanodiamonds in their cellular effects. The aim of this study was to check the ability of surface-modified detonation nanodiamonds to reach intracellular compartments without degradation of the surface-conjugated drug or fluorescent marker. In this study we examined the penetration HUVEC-ST and A549 cells by detonation nanodiamonds (grain size <20 nm) modified by adding to, employing four pharmacological inhibitors of endocytosis, using optical, confocal and transmission electron microscopy We discuss the possibilities, the challenges of studying the endocytic pathways involved in cellular uptake of nanoparticles. Our results suggest that fluorescent nanomaterials are very promising for monitoring the intracellular fate of nanodiamonds.
Collapse
|
163
|
Detection of vascular endothelial growth factor in colon cancer xenografts using bevacizumab based near infrared fluorophore conjugate. J Biomed Sci 2014; 21:35. [PMID: 24780003 PMCID: PMC4012715 DOI: 10.1186/1423-0127-21-35] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/23/2014] [Indexed: 01/06/2023] Open
Abstract
Background The aim of this study was to develop the near infrared fluorescence (NIRF)-based imaging agent for the visualization of vascular endothelial growth factor (VEGF) in colon cancer. AlexaFluor 750 conjugating with bevacizumab, and injected intravenously into nude mice bearing VEGF over-expressing HT29 human colorectal cancer. Optical imaging was performed at 15 min, 24 h and 48 h post injection. Immunofluorescences staining of the tumor sections were performed. HT29 colorectal cancer xenografts were clearly visualized with bevacizumab-AlexaFluor 750. Results Ex vivo analysis showed 2.1 ± 0.4%, 37.6 ± 6.3% and 38.5 ± 6.2% injected dose/g accumulated in the tumors at 15 min, 24 h and 48 h respectively. Tumor uptake was significantly decreased in pretreated with excess of bevacizumab (p = 0.002). Immunofluorescence analysis showed strong staining of anti-CD 31 antibody around the blood vessels. Anti-VEGF-A and bevacizumab showed heterogeneous expression throughout the tumor. Conclusions Current study successfully detected the VEGF expression in HT29 colorectal cancer xenografts, signifying as a potential agent for non-invasive imaging of VEGF expression, which may be applied in clinical practice.
Collapse
|
164
|
Heylman C, Sobrino A, Shirure VS, Hughes CC, George SC. A strategy for integrating essential three-dimensional microphysiological systems of human organs for realistic anticancer drug screening. Exp Biol Med (Maywood) 2014; 239:1240-54. [PMID: 24740872 DOI: 10.1177/1535370214525295] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality around the world. Despite some success, traditional anticancer drugs developed to reduce tumor growth face important limitations primarily due to undesirable bone marrow and cardiovascular toxicity. Many drugs fail in clinical development after showing promise in preclinical trials, suggesting that the available in vitro and animal models are poor predictors of drug efficacy and toxicity in humans. Thus, novel models that more accurately mimic the biology of human organs are necessary for high-throughput drug screening. Three-dimensional (3D) microphysiological systems can utilize induced pluripotent stem cell technology, tissue engineering, and microfabrication techniques to develop tissue models of human tumors, cardiac muscle, and bone marrow on the order of 1 mm(3) in size. A functional network of human capillaries and microvessels to overcome diffusion limitations in nutrient delivery and waste removal can also nourish the 3D microphysiological tissues. Importantly, the 3D microphysiological tissues are grown on optically clear platforms that offer non-invasive and non-destructive image acquisition with subcellular resolution in real time. Such systems offer a new paradigm for high-throughput drug screening and will significantly improve the efficiency of identifying new drugs for cancer treatment that minimize cardiac and bone marrow toxicity.
Collapse
Affiliation(s)
- Christopher Heylman
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA
| | - Agua Sobrino
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA
| | - Christopher Cw Hughes
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA Department of Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
165
|
Seo BR, DelNero P, Fischbach C. In vitro models of tumor vessels and matrix: engineering approaches to investigate transport limitations and drug delivery in cancer. Adv Drug Deliv Rev 2014; 69-70:205-216. [PMID: 24309015 DOI: 10.1016/j.addr.2013.11.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/14/2013] [Accepted: 11/24/2013] [Indexed: 12/12/2022]
Abstract
Tumor-stroma interactions have emerged as critical determinants of drug efficacy. However, the underlying biological and physicochemical mechanisms by which the microenvironment regulates therapeutic response remain unclear, due in part to a lack of physiologically relevant in vitro platforms to accurately interrogate tissue-level phenomena. Tissue-engineered tumor models are beginning to address this shortcoming. By allowing selective incorporation of microenvironmental complexity, these platforms afford unique access to tumor-associated signaling and transport dynamics. This review will focus on engineering approaches to study drug delivery as a function of tumor-associated changes of the vasculature and extracellular matrix (ECM). First, we review current biological understanding of these components and discuss their impact on transport processes. Then, we evaluate existing microfluidic, tissue engineering, and materials science strategies to recapitulate vascular and ECM characteristics of tumors, and finish by outlining challenges and future directions of the field that may ultimately improve anti-cancer therapies.
Collapse
|
166
|
Fleuren EDG, Versleijen-Jonkers YMH, Heskamp S, van Herpen CML, Oyen WJG, van der Graaf WTA, Boerman OC. Theranostic applications of antibodies in oncology. Mol Oncol 2014; 8:799-812. [PMID: 24725480 DOI: 10.1016/j.molonc.2014.03.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/10/2014] [Indexed: 02/07/2023] Open
Abstract
Targeted therapies, including antibodies, are becoming increasingly important in cancer therapy. Important limitations, however, are that not every patient benefits from a specific antibody therapy and that responses could be short-lived due to acquired resistance. In addition, targeted therapies are quite expensive and are not completely devoid of side-effects. This urges the need for accurate patient selection and response monitoring. An important step towards personalizing antibody treatment could be the implementation of theranostics. Antibody theranostics combine the diagnostic and therapeutic potential of an antibody, thereby selecting those patients who are most likely to benefit from antibody treatment. This review focuses on the clinical application of theranostic antibodies in oncology. It provides detailed information concerning the suitability of antibodies for theranostics, the different types of theranostic tests available and summarizes the efficacy of theranostic antibodies used in current clinical practice. Advanced theranostic applications, including radiolabeled antibodies for non-invasive functional imagining, are also addressed. Finally, we discuss the importance of theranostics in the emerging field of personalized medicine and critically evaluate recent data to determine the best way to apply antibody theranostics in the future.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | - Sandra Heskamp
- Department of Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Otto C Boerman
- Department of Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
167
|
Akashi Y, Oda T, Ohara Y, Miyamoto R, Kurokawa T, Hashimoto S, Enomoto T, Yamada K, Satake M, Ohkohchi N. Anticancer effects of gemcitabine are enhanced by co-administered iRGD peptide in murine pancreatic cancer models that overexpressed neuropilin-1. Br J Cancer 2014; 110:1481-7. [PMID: 24556620 PMCID: PMC3960621 DOI: 10.1038/bjc.2014.49] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/27/2013] [Accepted: 01/14/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Impaired drug transport is an important factor that reduces the efficacy of anticancer agents against pancreatic cancer. Here, we report a novel combination chemotherapy using gemcitabine (GEM) and internalised-RGD (iRGD) peptide, which enhances tumour-specific drug penetration by binding neuropilin-1 (NRP1) receptor. METHODS A total of five pancreatic cancer murine models (two cell line-based xenografts (CXs) and three tumour grafts (TGs)) were treated with either GEM (100 mg kg(-1), q3d × 4) alone or GEM plus iRGD peptide (8 μmol kg(-1)). Evaluation of NRP1 expression in xenografts and 48 clinical cancer specimens was performed by immunohistochemistry (IHC). RESULTS We identified a subset of pancreatic cancer models that showed NRP1 overexpression sensitive to iRGD co-administration. Treatment with GEM plus iRGD peptide resulted in a significant tumour reduction compared with GEM monotherapy in CXs, but not remarkable in TGs. Potential targets of iRGD were characterised as cases showing NRP1 overexpression (IHC-2+/3+), and these accounted for 45.8% of the clinical specimens. CONCLUSIONS Internalised RGD peptide enhances the effects of co-administered drugs in pancreatic cancer models, its efficacy is however only appreciable in those employing cell lines. Therefore, the clinical application needs to be given careful consideration.
Collapse
Affiliation(s)
- Y Akashi
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - T Oda
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - Y Ohara
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - R Miyamoto
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - T Kurokawa
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - S Hashimoto
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - T Enomoto
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - K Yamada
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| | - M Satake
- Department of Diagnostic Radiology, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - N Ohkohchi
- Department of Surgery, Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575 Ibaraki, Japan
| |
Collapse
|
168
|
Groh CM, Hubbard ME, Jones PF, Loadman PM, Periasamy N, Sleeman BD, Smye SW, Twelves CJ, Phillips RM. Mathematical and computational models of drug transport in tumours. J R Soc Interface 2014; 11:20131173. [PMID: 24621814 DOI: 10.1098/rsif.2013.1173] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The ability to predict how far a drug will penetrate into the tumour microenvironment within its pharmacokinetic (PK) lifespan would provide valuable information about therapeutic response. As the PK profile is directly related to the route and schedule of drug administration, an in silico tool that can predict the drug administration schedule that results in optimal drug delivery to tumours would streamline clinical trial design. This paper investigates the application of mathematical and computational modelling techniques to help improve our understanding of the fundamental mechanisms underlying drug delivery, and compares the performance of a simple model with more complex approaches. Three models of drug transport are developed, all based on the same drug binding model and parametrized by bespoke in vitro experiments. Their predictions, compared for a 'tumour cord' geometry, are qualitatively and quantitatively similar. We assess the effect of varying the PK profile of the supplied drug, and the binding affinity of the drug to tumour cells, on the concentration of drug reaching cells and the accumulated exposure of cells to drug at arbitrary distances from a supplying blood vessel. This is a contribution towards developing a useful drug transport modelling tool for informing strategies for the treatment of tumour cells which are 'pharmacokinetically resistant' to chemotherapeutic strategies.
Collapse
Affiliation(s)
- C M Groh
- Klinik und Poliklinik für Strahlentherapie (Medizinische Physik), Universitätsklinikum Würzburg, , Josef-Schneider-Strasse 11, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Munson J, Bonner M, Fried L, Hofmekler J, Arbiser J, Bellamkonda R. Identifying new small molecule anti-invasive compounds for glioma treatment. Cell Cycle 2014; 12:2200-9. [PMID: 24067366 DOI: 10.4161/cc.25334] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma is a disease with poor survival rates after diagnosis. Treatment of the disease involves debulking of the tumor, which is limited by the degree of invasiveness of the disease. Therefore, a treatment to halt the invasion of glioma is desirable for clinical implementation. There have been several candidate compounds targeting specific aspects of invasion, including cell adhesions, matrix degradation, and cytoskeletal rearrangement, but they have failed clinically for a variety of reasons. New targets against glioma invasion include upstream mediators of these classical targets in an effort to better inhibit invasion with more specificity for cancer. Included in these treatments is a new class of compounds inhibiting the generation of reactive oxygen species by targeting the NADPH oxidases. These compounds stand to inhibit multiple pathways, including nuclear factor kappa B and Akt. By conducting a screen of compounds thought to inhibit these pathways, a new compound to halt invasion was found that may have a beneficial effect against glioma, based on recent publications. Further, there are still limitations to the treatment of glioblastoma regardless of the discovery of new targets and compounds that should be addressed to better the therapies against this deadly cancer.
Collapse
Affiliation(s)
- Jennifer Munson
- Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology; Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
170
|
Samanta A, Jana S, Das RK, Chang YT. Biocompatible surface-enhanced Raman scattering nanotags for in vivo cancer detection. Nanomedicine (Lond) 2014; 9:523-35. [DOI: 10.2217/nnm.13.222] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The advancement of surface-enhanced Raman scattering (SERS) is significantly increasing as an ultra-sensitive sensing technology in biomedical research. In this review, we focus on the most recent developments of biocompatible nanoprobes for cancer research. First, we discuss coating approaches to enhance the biocompatibility of SERS substrate and Raman reporters. Furthermore, interesting ligands such as antibodies, aptamers and polypeptides are attached to the surface of nanotags for targeting the cancerous cells in vitro. The unique multiplexing capabilities of the SERS technique have been applied for simultaneous multiple target recognition. Finally, these noninvasive, ultrasensitive tools are mostly highlighted for in vivo tumor detection. Potential application of SERS nanotags in therapeutic study and the possibility of SERS nanotags in biomedical applications are outlined briefly in this review.
Collapse
Affiliation(s)
- Animesh Samanta
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A *STAR), 11 Biopolis Way, 138667, Singapore
| | - Santanu Jana
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A *STAR), 11 Biopolis Way, 138667, Singapore
| | - Raj Kumar Das
- Department of Chemistry & MedChem Program for Life Sciences, National University of Singapore, 3 Science Drive 3, 117543, Singapore
| | - Young Tae Chang
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A *STAR), 11 Biopolis Way, 138667, Singapore
- Department of Chemistry & MedChem Program for Life Sciences, National University of Singapore, 3 Science Drive 3, 117543, Singapore
| |
Collapse
|
171
|
Fleuren EDG, Versleijen-Jonkers YMH, Boerman OC, van der Graaf WTA. Targeting receptor tyrosine kinases in osteosarcoma and Ewing sarcoma: current hurdles and future perspectives. Biochim Biophys Acta Rev Cancer 2014; 1845:266-76. [PMID: 24582852 DOI: 10.1016/j.bbcan.2014.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/20/2014] [Accepted: 02/22/2014] [Indexed: 12/26/2022]
Abstract
Osteosarcoma (OS) and Ewing sarcoma (ES) are the two most common types of primary bone cancer, which mainly affect children and young adults. Despite intensive multi-modal treatment, the survival of both OS and ES has not improved much during the last decades and new therapeutic options are awaited. One promising approach is the specific targeting of transmembrane receptor tyrosine kinases (RTKs) implicated in these types of bone cancer. However, despite encouraging in vitro and in vivo results, apart from intriguing results of Insulin-like Growth Factor-1 Receptor (IGF-1R) antibodies in ES, clinical studies are limited or disappointing. Primary resistance to RTK inhibitors is frequently observed in OS and ES patients, and even patients that initially respond well eventually develop acquired resistance. There are, however, a few remarks to make concerning the current set-up of clinical trials and about strategies to improve RTK-based treatments in OS and ES. This review provides an overview concerning current RTK-mediated therapies in OS and ES and discusses the problems observed in the clinic. More importantly, we describe several strategies to overcome resistance to RTK inhibitors which may significantly improve outcome of OS and ES patients.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | - Otto C Boerman
- Department of Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
172
|
Thurber GM, Reiner T, Yang KS, Kohler RH, Weissleder R. Effect of small-molecule modification on single-cell pharmacokinetics of PARP inhibitors. Mol Cancer Ther 2014; 13:986-95. [PMID: 24552776 DOI: 10.1158/1535-7163.mct-13-0801] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heterogeneous delivery of drugs in tumors is an established process contributing to variability in treatment outcome. Despite the general acceptance of variable delivery, the study of the underlying causes is challenging, given the complex tumor microenvironment including intra- and intertumor heterogeneity. The difficulty in studying this distribution is even more significant for small-molecule drugs where radiolabeled compounds or mass spectrometry detection lack the spatial and temporal resolution required to quantify the kinetics of drug distribution in vivo. In this work, we take advantage of the synthesis of fluorescent drug conjugates that retain their target binding but are designed with different physiochemical and thus pharmacokinetic properties. Using these probes, we followed the drug distribution in cell culture and tumor xenografts with temporal resolution of seconds and subcellular spatial resolution. These measurements, including in vivo permeability of small-molecule drugs, can be used directly in predictive pharmacokinetic models for the design of therapeutics and companion imaging agents as demonstrated by a finite element model.
Collapse
Affiliation(s)
- Greg M Thurber
- Authors' Affiliations: Center for Systems Biology, Massachusetts General Hospital; and Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
173
|
Wu W, Driessen W, Jiang X. Oligo(ethylene glycol)-Based Thermosensitive Dendrimers and Their Tumor Accumulation and Penetration. J Am Chem Soc 2014; 136:3145-55. [DOI: 10.1021/ja411457r] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wei Wu
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Provincial Laboratory for Nanotechnology, Nanjing University, Nanjing 210093, People’s Republic of China
| | | | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Provincial Laboratory for Nanotechnology, Nanjing University, Nanjing 210093, People’s Republic of China
| |
Collapse
|
174
|
Liu Z, Yang Z, Jiang S, Zou Q, Yuan Y, Li J, Li D, Liang L, Chen M, Chen S. Paxillin and carbonic anhydrase IX are prognostic markers in gallbladder squamous cell/adenosquamous carcinomas and adenocarcinomas. Histopathology 2014; 64:921-34. [PMID: 24354963 DOI: 10.1111/his.12341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 11/26/2013] [Accepted: 12/02/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Ziru Liu
- Research Laboratory of Hepatobiliary Diseases; Second Xiangya Hospital; Central South University; Changsha China
| | - Zhulin Yang
- Research Laboratory of Hepatobiliary Diseases; Second Xiangya Hospital; Central South University; Changsha China
| | - Song Jiang
- Research Laboratory of Hepatobiliary Diseases; Second Xiangya Hospital; Central South University; Changsha China
| | - Qiong Zou
- Department of Pathology; Third Xiangya Hospital; Central South University; Changsha China
| | - Yuan Yuan
- Department of Pathology; Third Xiangya Hospital; Central South University; Changsha China
| | - Jinghe Li
- Department of Pathology; Basic School of Medicine; Changsha China
| | - Daiqiang Li
- Department of Pathology; Second Xiangya Hospital; Central South University; Changsha China
| | - Lufeng Liang
- Department of Hepatobiliary and Pancreatic Surgery; Hunan Provincial People's Hospital; Changsha China
| | - Meigui Chen
- Department of Pathology; Loudi Central Hospital; Loudi China
| | - Senlin Chen
- Department of Pathology; Hunan Provincial Tumor Hospital; Changsha China
| |
Collapse
|
175
|
Gao H, Yang Z, Cao S, Xiong Y, Zhang S, Pang Z, Jiang X. Tumor cells and neovasculature dual targeting delivery for glioblastoma treatment. Biomaterials 2014; 35:2374-82. [DOI: 10.1016/j.biomaterials.2013.11.076] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 11/25/2013] [Indexed: 01/07/2023]
|
176
|
Abstract
Cancer nanotechnology is currently under intense development for applications in cancer imaging, molecular diagnosis and targeted therapy. The basic rationale is that nanometer-sized particles, such as biodegradable micelles, semiconductor quantum dots and iron oxide nanocrystals, have functional or structural properties that are not available from either molecular or macroscopic agents. When linked with biotargeting ligands, such as monoclonal antibodies, peptides or small molecules, these nanoparticles are used to target malignant tumors with high affinity and specificity. In the 'mesoscopic' size range of 5-100 nm in diameter, nanoparticles also have large surface areas and functional groups for conjugating to multiple diagnostic (e.g., optical, radioisotopic or magnetic) and therapeutic (e.g., anticancer) agents. Recent advances have led to multifunctional nanoparticle probes for molecular and cellular imaging, nanoparticle drugs for targeted therapy, and integrated nanodevices for early cancer detection and screening. These developments have opened exciting opportunities for personalized oncology in which cancer detection, diagnosis and therapy are tailored to each individual's molecular profile, and also for predictive oncology, in which genetic/molecular information is used to predict tumor development, progression and clinical outcome.
Collapse
Affiliation(s)
- May D Wang
- Georgia Institute of Technology, Department of Biomedical Engineering, and Emory University, Atlanta, GA 30332, USA.
| | | | | | | |
Collapse
|
177
|
Heskamp S, van Laarhoven HWM, van der Graaf WTA, Oyen WJG, Boerman OC. Radionuclide imaging of drug delivery for patient selection in targeted therapy. Expert Opin Drug Deliv 2014; 11:175-85. [DOI: 10.1517/17425247.2014.870552] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
178
|
Pawar PV, Domb AJ, Kumar N. Systemic Targeting Systems-EPR Effect, Ligand Targeting Systems. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2014. [DOI: 10.1007/978-1-4614-9434-8_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
179
|
Bioinformatics and Nanotechnologies: Nanomedicine. SPRINGER HANDBOOK OF BIO-/NEUROINFORMATICS 2014. [PMCID: PMC7124100 DOI: 10.1007/978-3-642-30574-0_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this chapter we focus on the bioinformatics strategies for translating genome-wide expression analyses into clinically useful cancer markers
with a specific focus on breast cancer
with a perspective on new diagnostic device tools coming from the field of nanobiotechnology and the challenges related to high-throughput data integration, analysis, and assessment from multiple sources. Great progress in the development of molecular biology techniques has been seen since the discovery of the structure of deoxyribonucleic acid (DNA) and the implementation of a polymerase chain reaction (PCR) method. This started a new era of research on the structure of nucleic acids molecules, the development of new analytical tools, and DNA-based analyses that allowed the sequencing of the human genome, the completion of which has led to intensified efforts toward comprehensive analysis of mammalian cell struc ture and metabolism in order to better understand the mechanisms that regulate normal cell behavior and identify the gene alterations responsible for a broad spectrum of human diseases, such as cancer, diabetes, cardiovascular diseases, neurodegenerative disorders, and others.
Collapse
|
180
|
Upponi JR, Torchilin VP. Passive vs. Active Targeting: An Update of the EPR Role in Drug Delivery to Tumors. NANO-ONCOLOGICALS 2014. [DOI: 10.1007/978-3-319-08084-0_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
181
|
Cattaneo L, Zunino P. Computational models for fluid exchange between microcirculation and tissue interstitium. ACTA ACUST UNITED AC 2014. [DOI: 10.3934/nhm.2014.9.135] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
182
|
Jang BS. MicroSPECT and MicroPET Imaging of Small Animals for Drug Development. Toxicol Res 2013; 29:1-6. [PMID: 24278622 PMCID: PMC3834443 DOI: 10.5487/tr.2013.29.1.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 03/14/2013] [Accepted: 03/18/2013] [Indexed: 11/21/2022] Open
Abstract
The process of drug discovery and development requires substantial resources and time. The drug industry has tried to reduce costs by conducting appropriate animal studies together with molecular biological and genetic analyses. Basic science research has been limited to in vitro studies of cellular processes and ex vivo tissue examination using suitable animal models of disease. However, in the past two decades new technologies have been developed that permit the imaging of live animals using radiotracer emission, Xrays, magnetic resonance signals, fluorescence, and bioluminescence. The main objective of this review is to provide an overview of small animal molecular imaging, with a focus on nuclear imaging (single photon emission computed tomography and positron emission tomography). These technologies permit visualization of toxicodynamics as well as toxicity to specific organs by directly monitoring drug accumulation and assessing physiological and/or molecular alterations. Nuclear imaging technology has great potential for improving the efficiency of the drug development process.
Collapse
Affiliation(s)
- Beom-Su Jang
- RI-Biomics Research & Development Team, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeonbuk, Korea
| |
Collapse
|
183
|
Kirui DK, Koay EJ, Guo X, Cristini V, Shen H, Ferrari M. Tumor vascular permeabilization using localized mild hyperthermia to improve macromolecule transport. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:1487-96. [PMID: 24262998 DOI: 10.1016/j.nano.2013.11.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 10/04/2013] [Accepted: 11/05/2013] [Indexed: 01/14/2023]
Abstract
The abnormal tumor vasculature presents a major challenge to the adequate delivery of chemotherapeutics, often limiting efficacy. We developed a nanoparticle-based technique to deliver localized mild hyperthermia (MHT) used to transiently alter tumor vascular transport properties and enhance transport of macromolecules into tumor interstitium. The strategy involved administering and localizing accumulation of stealth gold nanorods (GNRs, 103 μg of GNRs/g of tumor), and irradiating tumor with a low-photon laser flux (1 W/cm(2)) to generate MHT. The treatment increased vascular permeability within 24 h after treatment, allowing enhanced transport of macromolecules up to 54 nm in size. A mathematical model is used to describe changes in tumor mass transport properties where the rate of macromolecular exchange between interstitial and vascular region (R) and maximum dye enhancement (Ymax) of 23-nm dextran dye is analytically solved. During enhanced permeability, R increased by 200% while Ymax increased by 30% relative to untreated group in pancreatic CAPAN-1 tumors. MHT treatment also enhanced transport of larger dextran dye (54 nm) as assessed by intravital microscopy, without causing occlusive cellular damage. Enhanced vascular transport was prolonged for up to 24 h after treatment, but reversible with transport parameters returning to basal levels after 36 h. This study indicates that localized mild hyperthermia treatment opens a transient time-window with which to enable and augment macromolecule transport and potentially improve therapeutic efficacy. From the clinical editor: In this study, local intra-tumor mild hyperthermia is induced using a nanoparticle-based approach utilizing stealth gold nanorods and irradiating the tumor with low-photon laser flux, resulting in locally increased vascular permeability enabling enhanced delivery of therapeutics, including macromolecules up to 54 nm in size. Similar approaches would be very helpful in addressing treatment-resistant malignancies in clinical practice.
Collapse
Affiliation(s)
| | - Eugene J Koay
- Houston Methodist Research Institute, Houston, TX, USA; MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaojing Guo
- Houston Methodist Research Institute, Houston, TX, USA; Department of Breast Cancer Pathology and Research Laboratory, Key Laboratory of Breast Cancer of Breast Cancer Prevention and Therapy, Tianjin, China
| | - Vittorio Cristini
- Department of Chemical and Nuclear Engineering, Center for Biomedical Engineering, The University of New Mexico, Albuquerque, NM, USA
| | - Haifa Shen
- Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medical College, New York, NY, USA
| | - Mauro Ferrari
- Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
184
|
Gao Y, Xie J, Chen H, Gu S, Zhao R, Shao J, Jia L. Nanotechnology-based intelligent drug design for cancer metastasis treatment. Biotechnol Adv 2013; 32:761-77. [PMID: 24211475 DOI: 10.1016/j.biotechadv.2013.10.013] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 10/19/2013] [Accepted: 10/29/2013] [Indexed: 12/21/2022]
Abstract
Traditional chemotherapy used today at clinics is mainly inherited from the thinking and designs made four decades ago when the Cancer War was declared. The potency of those chemotherapy drugs on in-vitro cancer cells is clearly demonstrated at even nanomolar levels. However, due to their non-specific effects in the body on normal tissues, these drugs cause toxicity, deteriorate patient's life quality, weaken the host immunosurveillance system, and result in an irreversible damage to human's own recovery power. Owing to their unique physical and biological properties, nanotechnology-based chemotherapies seem to have an ability to specifically and safely reach tumor foci with enhanced efficacy and low toxicity. Herein, we comprehensively examine the current nanotechnology-based pharmaceutical platforms and strategies for intelligent design of new nanomedicines based on targeted drug delivery system (TDDS) for cancer metastasis treatment, analyze the pros and cons of nanomedicines versus traditional chemotherapy, and evaluate the importance that nanomaterials can bring in to significantly improve cancer metastasis treatment.
Collapse
Affiliation(s)
- Yu Gao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingjing Xie
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Haijun Chen
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China; Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Fuzhou University, Fujian 350108, China
| | - Songen Gu
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Rongli Zhao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China.
| |
Collapse
|
185
|
Exploiting nanotechnology to overcome tumor drug resistance: Challenges and opportunities. Adv Drug Deliv Rev 2013; 65:1731-47. [PMID: 24036273 DOI: 10.1016/j.addr.2013.09.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/15/2013] [Accepted: 09/03/2013] [Indexed: 02/06/2023]
Abstract
Tumor cells develop resistance to chemotherapeutic drugs through multiple mechanisms. Overexpression of efflux transporters is an important source of drug resistance. Efflux transporters such as P-glycoprotein reduce intracellular drug accumulation and compromise drug efficacy. Various nanoparticle-based approaches have been investigated to overcome efflux-mediated resistance. These include the use of formulation excipients that inhibit transporter activity and co-delivery of the anticancer drug with a specific inhibitor of transporter function or expression. However, the effectiveness of nanoparticles can be diminished by poor transport in the tumor tissue. Hence, adjunct therapies that improve the intratumoral distribution of nanoparticles may be vital to the successful application of nanotechnology to overcome tumor drug resistance. This review discusses the mechanisms of tumor drug resistance and highlights the opportunities and challenges in the use of nanoparticles to improve the efficacy of anticancer drugs against resistant tumors.
Collapse
|
186
|
Chen H, Wang L, Yu Q, Qian W, Tiwari D, Yi H, Wang AY, Huang J, Yang L, Mao H. Anti-HER2 antibody and ScFvEGFR-conjugated antifouling magnetic iron oxide nanoparticles for targeting and magnetic resonance imaging of breast cancer. Int J Nanomedicine 2013; 8:3781-94. [PMID: 24124366 PMCID: PMC3794963 DOI: 10.2147/ijn.s49069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Antifouling magnetic iron oxide nanoparticles (IONPs) coated with block copolymer poly(ethylene oxide)-block-poly(γ-methacryloxypropyltrimethoxysilane) (PEO-b-PγMPS) were investigated for improving cell targeting by reducing nonspecific uptake. Conjugation of a HER2 antibody, Herceptin®, or a single chain fragment (ScFv) of antibody against epidermal growth factor receptor (ScFvEGFR) to PEO-b-PγMPS-coated IONPs resulted in HER2-targeted or EGFR-targeted IONPs (anti-HER2-IONPs or ScFvEGFR-IONPs). The anti-HER2-IONPs bound specifically to SK-BR-3, a HER2-overexpressing breast cancer cell line, but not to MDA-MB-231, a HER2-underexpressing cell line. On the other hand, the ScFvEGFR-IONPs showed strong reactivity with MDA-MB-231, an EGFR-positive human breast cancer cell line, but not with MDA-MB-453, an EGFR-negative human breast cancer cell line. Transmission electron microscopy revealed internalization of the receptor-targeted nanoparticles by the targeted cancer cells. In addition, both antibody-conjugated and non-antibody-conjugated IONPs showed reduced nonspecific uptake by RAW264.7 mouse macrophages in vitro. The developed IONPs showed a long blood circulation time (serum half-life 11.6 hours) in mice and low accumulation in both the liver and spleen. At 24 hours after systemic administration of ScFvEGFR-IONPs into mice bearing EGFR-positive breast cancer 4T1 mouse mammary tumors, magnetic resonance imaging revealed signal reduction in the tumor as a result of the accumulation of the targeted IONPs.
Collapse
Affiliation(s)
- Hongwei Chen
- Department of Radiology and Imaging Sciences, Atlanta, GA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Teo BM, van der Westen R, Hosta-Rigau L, Städler B. Cell response to PEGylated poly(dopamine) coated liposomes considering shear stress. Biochim Biophys Acta Gen Subj 2013; 1830:4838-47. [DOI: 10.1016/j.bbagen.2013.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
|
188
|
Chapman S, Dobrovolskaia M, Farahani K, Goodwin A, Joshi A, Lee H, Meade T, Pomper M, Ptak K, Rao J, Singh R, Sridhar S, Stern S, Wang A, Weaver JB, Woloschak G, Yang L. Nanoparticles for cancer imaging: The good, the bad, and the promise. NANO TODAY 2013; 8:454-460. [PMID: 25419228 PMCID: PMC4240321 DOI: 10.1016/j.nantod.2013.06.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent advances in molecular imaging and nanotechnology are providing new opportunities for biomedical imaging with great promise for the development of novel imaging agents. The unique optical, magnetic, and chemical properties of materials at the scale of nanometers allow the creation of imaging probes with better contrast enhancement, increased sensitivity, controlled biodistribution, better spatial and temporal information, multi-functionality and multi-modal imaging across MRI, PET, SPECT, and ultrasound. These features could ultimately translate to clinical advantages such as earlier detection, real time assessment of disease progression and personalized medicine. However, several years of investigation into the application of these materials to cancer research has revealed challenges that have delayed the successful application of these agents to the field of biomedical imaging. Understanding these challenges is critical to take full advantage of the benefits offered by nano-sized imaging agents. Therefore, this article presents the lessons learned and challenges encountered by a group of leading researchers in this field, and suggests ways forward to develop nanoparticle probes for cancer imaging. Published by Elsevier Ltd.
Collapse
Affiliation(s)
- Sandra Chapman
- Office of Cancer Nanotechnology Research, Center for Strategic Scientific Initiatives, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Marina Dobrovolskaia
- Nanotechnology Characterization Laboratory, SAIC-Frederick Inc., Advanced Technology Research Facility — Frederick National Laboratory for Cancer Research, P.O. Box B, Frederick, MD 21702, United States
| | - Keyvan Farahani
- Image-Guided Interventions Branch, Cancer Imaging Program, National Cancer Institute, Rockville, MD 20852, United States
| | - Andrew Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States
| | - Amit Joshi
- Department of Radiology, TBMM Graduate Program, Baylor College of Medicine, Houston, TX 77030, United States
- Department of Molecular Physiology and Biophysics, TBMM Graduate Program, Baylor College of Medicine, Houston, TX 77030, United States
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Thomas Meade
- Departments of Chemistry, Molecular Biosciences and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Martin Pomper
- Image-Guided Interventions Branch, Cancer Imaging Program, National Cancer Institute, Rockville, MD 20852, United States
| | - Krzysztof Ptak
- Johns Hopkins Medical School, Baltimore, MD 21287, United States
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305-5484, United States
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Srinivas Sridhar
- Nanomedicine Science and Technology Center and Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Stephan Stern
- Nanotechnology Characterization Laboratory, SAIC-Frederick Inc., Advanced Technology Research Facility — Frederick National Laboratory for Cancer Research, P.O. Box B, Frederick, MD 21702, United States
| | - Andrew Wang
- Department of Radiation Oncology, Lineberger Comprehensive Cancer, Carolina Center for Nanotechnology Excellence, University of North Carolina-Chapel Hill, NC 27599, United States
| | - John B. Weaver
- Department of Radiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, United States
| | - Gayle Woloschak
- Department of Radiation Oncology, Robert E. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, United States
- Department of Radiology, Robert E. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, United States
- Department of Cell and Molecular Biology, Robert E. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, United States
| |
Collapse
|
189
|
111In-cetuximab-F(ab')2 SPECT imaging for quantification of accessible epidermal growth factor receptors (EGFR) in HNSCC xenografts. Radiother Oncol 2013; 108:484-8. [PMID: 23932156 DOI: 10.1016/j.radonc.2013.06.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND PURPOSE Immunohistochemical epidermal growth factor receptor (EGFR) expression does not correlate with treatment response in head and neck squamous cell carcinomas (HNSCC). Aim was to apply the tracer (111)In-cetuximab-F(ab')2 for EGFR microSPECT imaging and to investigate if tracer uptake correlated with response to EGFR-inhibition by cetuximab in HNSCC xenografts. Usage of F(ab)2 fragments allows for shorter interval between tracer injection and imaging. MATERIALS AND METHODS Mice with HNSCC xenografts, SCCNij202, 153, 185 and 167 were imaged with microSPECT using (111)In-cetuximab-F(ab')2. Subsequently, tumors were analyzed by autoradiography and immunohistochemistry and tracer concentration was determined. Tumor uptake was correlated with previously assessed response to cetuximab treatment. RESULTS MicroSPECT imaging showed preferential uptake in HNSCC xenografts. Tumor-to-liver ratios were 3.1 ± 0.2 (SCCNij202), 2.8 ± 0.4 (SCCNij153), 2.0 ± 0.8 (SCCNij185), 2.0 ± 0.4 (SCCNij167). Immunohistochemical EGFR fractions (fEGFR) differed significantly between xenografts; 0.77 ± 0.07 (SCCNij202), 0.66 ± 0.11 (SCCNij153), 0.57 ± 0.19 (SCCNij185), 0.16 ± 0.10 (SCCNij167) (p < 0.001). Tumor fEGFR correlated with (111)In-cetuximab-F(ab')2 tumor uptake (r = 0.6, p < 0.01) and tracer autoradiography (r = 0.7, p < 0.0001). Tumor uptake of (111)In-cetuximab-F(ab')2 was proportionally associated with cetuximab treatment response in three out of four xenograft models. CONCLUSION (111)In-cetuximab-F(ab')2 showed good tumor-to-background contrast on microSPECT imaging, allowing noninvasive assessment of EGFR expression in vivo, and possibly evaluation of treatment response to EGFR-inhibition.
Collapse
|
190
|
Interstitial fluid flow and drug delivery in vascularized tumors: a computational model. PLoS One 2013; 8:e70395. [PMID: 23940570 PMCID: PMC3734291 DOI: 10.1371/journal.pone.0070395] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/18/2013] [Indexed: 01/15/2023] Open
Abstract
Interstitial fluid is a solution that bathes and surrounds the human cells and provides them with nutrients and a way of waste removal. It is generally believed that elevated tumor interstitial fluid pressure (IFP) is partly responsible for the poor penetration and distribution of therapeutic agents in solid tumors, but the complex interplay of extravasation, permeabilities, vascular heterogeneities and diffusive and convective drug transport remains poorly understood. Here we consider-with the help of a theoretical model-the tumor IFP, interstitial fluid flow (IFF) and its impact upon drug delivery within tumor depending on biophysical determinants such as vessel network morphology, permeabilities and diffusive vs. convective transport. We developed a vascular tumor growth model, including vessel co-option, regression, and angiogenesis, that we extend here by the interstitium (represented by a porous medium obeying Darcy's law) and sources (vessels) and sinks (lymphatics) for IFF. With it we compute the spatial variation of the IFP and IFF and determine its correlation with the vascular network morphology and physiological parameters like vessel wall permeability, tissue conductivity, distribution of lymphatics etc. We find that an increased vascular wall conductivity together with a reduction of lymph function leads to increased tumor IFP, but also that the latter does not necessarily imply a decreased extravasation rate: Generally the IF flow rate is positively correlated with the various conductivities in the system. The IFF field is then used to determine the drug distribution after an injection via a convection diffusion reaction equation for intra- and extracellular concentrations with parameters guided by experimental data for the drug Doxorubicin. We observe that the interplay of convective and diffusive drug transport can lead to quite unexpected effects in the presence of a heterogeneous, compartmentalized vasculature. Finally we discuss various strategies to increase drug exposure time of tumor cells.
Collapse
|
191
|
Pastorino F, Brignole C, Loi M, Di Paolo D, Di Fiore A, Perri P, Pagnan G, Ponzoni M. Nanocarrier-mediated targeting of tumor and tumor vascular cells improves uptake and penetration of drugs into neuroblastoma. Front Oncol 2013; 3:190. [PMID: 23936762 PMCID: PMC3733002 DOI: 10.3389/fonc.2013.00190] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/08/2013] [Indexed: 11/15/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children, accounting for about 8% of childhood cancers. Despite aggressive treatment, patients suffering from high-risk NB have very poor 5-year overall survival rate, due to relapsed and/or treatment-resistant tumors. A further increase in therapeutic dose intensity is not feasible, because it will lead to prohibitive short-term and long-term toxicities. New approaches with targeted therapies may improve efficacy and decrease toxicity. The use of drug delivery systems allows site specific delivery of higher payload of active agents associated with lower systemic toxicity compared to the use of conventional (“free”) drugs. The possibility of imparting selectivity to the carriers to the cancer foci through the use of a targeting moiety (e.g., a peptide or an antibody) further enhances drug efficacy and safety. We have recently developed two strategies for increasing local concentration of anti-cancer agents, such as CpG-containing oligonucleotides, small interfering RNAs, and chemotherapeutics in NB. For doing that, we have used the monoclonal antibody anti-disialoganglioside (GD2), able to specifically recognize the NB tumor and the peptides containing NGR and CPRECES motifs, that selectively bind to the aminopeptidase N-expressing endothelial and the aminopeptidase A-expressing perivascular tumor cells, respectively. The review will focus on the use of tumor- and tumor vasculature-targeted nanocarriers to improve tumor targeting, uptake, and penetration of drugs in preclinical models of human NB.
Collapse
Affiliation(s)
- Fabio Pastorino
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini , Genoa , Italy
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Fu X, Rivera A, Tao L, Zhang X. Genetically modified T cells targeting neovasculature efficiently destroy tumor blood vessels, shrink established solid tumors and increase nanoparticle delivery. Int J Cancer 2013; 133:2483-92. [PMID: 23661285 DOI: 10.1002/ijc.28269] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/23/2013] [Indexed: 02/03/2023]
Abstract
Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvβ3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvβ3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX
| | | | | | | |
Collapse
|
193
|
Hao G, Kumar A, Dobin T, Oz OK, Hsieh JT, Sun X. A multivalent approach of imaging probe design to overcome an endogenous anion binding competition for noninvasive assessment of prostate specific membrane antigen. Mol Pharm 2013; 10:2975-85. [PMID: 23768233 DOI: 10.1021/mp4000844] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
2[(3-Amino-3-carboxypropyl)(hydroxy)(phosphinyl)methyl]pentane-1,5-dioic acid) (GPI) is a highly potent inhibitor of prostate specific membrane antigen (PSMA) with a rapid in vivo clearance profile from nontarget organs including kidneys, but its use for imaging of PSMA is impeded by an endogenous anion (serum phosphate) competition, which compromises its specific binding to the antigen. Multipresentation of a targeting molecule on a single entity has been recognized as a practical way for imaging sensitivity enhancement. Herein, we demonstrate a multivalent approach based on a (64)Cu-specific bifunctional chelator scaffold to overcome the endogenous phosphate competition thus enabling the utility of GPI conjugates for in vivo detection of PSMA and imaging quantification. Both monomeric (H2CBT1G) and dimeric (H2CBT2G) conjugates were synthesized and labeled with (64)Cu for in vitro and in vivo evaluations. A 4-fold enhancement of PSMA binding affinity was observed for H2CBT2G as compared to H2CBT1G from the PSMA competitive binding assays performed on LNCaP cells. In vivo PET imaging studies were conducted on mouse xenograft models established with a PSMA(+) cell line, LNCaP, and PSMA(-) PC3 and H2009 cell lines. (64)Cu-CBT2G showed significantly higher LNCaP tumor uptake than (64)Cu-CBT1G at 1, 4, and 24 h postinjection (p.i.) (p < 0.05). In addition, tumor uptake of (64)Cu-CBT2G remained steady out to 24 h p.i. (1.46 ± 0.54, 1.12 ± 0.56, and 1.00 ± 0.50% ID/g at 1, 4, and 24 h p.i., respectively), while (64)Cu-CBT1G showed a great decrease from 1 to 4 h p.i. The PSMA imaging specificity of both H2CBT1G and H2CBT2G was demonstrated by their low uptake in PSMA(-) tumors (PC3 and H2009) and further confirmed by a significant signal reduction in PSMA(+) LNCaP tumors in the blockade study. In addition, the LNCaP tumor uptake (% ID/g) of (64)Cu-CBT2G was found to be in a positive linear correlation with the tumor size (R(2) = 0.92, 0.94, and 0.93 for 1 h, 4 h, and 24 h p.i.). This may render the probe with potential application in the management of patients with prostate cancer.
Collapse
Affiliation(s)
- Guiyang Hao
- Department of Radiology, The University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | | | | | | | | | | |
Collapse
|
194
|
Shin SJ, Beech JR, Kelly KA. Targeted nanoparticles in imaging: paving the way for personalized medicine in the battle against cancer. Integr Biol (Camb) 2013; 5:29-42. [PMID: 22790418 DOI: 10.1039/c2ib20047c] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The way we view cancer has advanced greatly in the past few decades from simplistic approaches to finely honed systems. This transition has been made possible because of advancements on two fronts: the first is the rapidly expanding knowledge base of the mechanisms and characteristics of cancer; the second is innovation in imaging agent design. Rapid advancements in imaging and therapeutic agents are being made through the evolution from one-dimensional molecules to multi-functional nanoparticles. Powerful new agents that have high specificity and minimal toxicity are being developed for in vivo imaging. Here we detail the unique characteristics of cancer that allow differentiation from normal tissue and how they are exploited in nanoparticle imaging development. Firstly, genetic alterations, either endogenous or induced through gene therapy, are one such class of characteristics. Proteomic differences such as overexpressed surface receptors is another targetable feature used for enhanced nanoparticle retention. Increased need for nutrients and specific growth signals to sustain proliferation and angiogenesis are further examples of how cancer can be targeted. Lastly, migration and invasion through a unique microenvironment are two additional traits that are exploitable, due to differences in metalloproteinase concentrations and other factors. These differences are guiding current nanoparticle design to better target, image and treat cancer.
Collapse
Affiliation(s)
- Soo J Shin
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Sciences, PO Box 800759, Health System, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
195
|
Wei T, Liu J, Ma H, Cheng Q, Huang Y, Zhao J, Huo S, Xue X, Liang Z, Liang XJ. Functionalized nanoscale micelles improve drug delivery for cancer therapy in vitro and in vivo. NANO LETTERS 2013; 13:2528-34. [PMID: 23634882 DOI: 10.1021/nl400586t] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Poor penetration of therapeutic drugs into tumors is a major challenge in anticancer therapy, especially in solid tumors, leading to reduced therapeutic efficacy in vivo. In the study, we used a new tumor-penetrating peptide, CRGDK, to conjugate onto the surface of doxorubicin encapsulated nanoscale micelles. The CRGDK peptide triggered specific binding to neuropilin-1, leading to enhanced cellular uptake and cytotoxicity in vitro and highly accumulation and penetration in the tumors in vivo.
Collapse
Affiliation(s)
- Tuo Wei
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Hosta-Rigau L, Städler B. Shear Stress and Its Effect on the Interaction of Myoblast Cells with Nanosized Drug Delivery Vehicles. Mol Pharm 2013; 10:2707-12. [DOI: 10.1021/mp4001298] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Brigitte Städler
- iNANO Interdisciplinary
Nanoscience Centre, Aarhus
University, Denmark
| |
Collapse
|
197
|
Fang M, Peng CW, Pang DW, Li Y. Quantum dots for cancer research: current status, remaining issues, and future perspectives. Cancer Biol Med 2013; 9:151-63. [PMID: 23691472 PMCID: PMC3643664 DOI: 10.7497/j.issn.2095-3941.2012.03.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/19/2012] [Indexed: 12/31/2022] Open
Abstract
Cancer is a major threat to public health in the 21st century because it is one of the leading causes of death worldwide. The mechanisms of carcinogenesis, cancer invasion, and metastasis remain unclear. Thus, the development of a novel approach for cancer detection is urgent, and real-time monitoring is crucial in revealing its underlying biological mechanisms. With the optical and chemical advantages of quantum dots (QDs), QD-based nanotechnology is helpful in constructing a biomedical imaging platform for cancer behavior study. This review mainly focuses on the application of QD-based nanotechnology in cancer cell imaging and tumor microenvironment studies both in vivo and in vitro, as well as the remaining issues and future perspectives.
Collapse
Affiliation(s)
- Min Fang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan 430071, China
| | | | | | | |
Collapse
|
198
|
Cassette E, Helle M, Bezdetnaya L, Marchal F, Dubertret B, Pons T. Design of new quantum dot materials for deep tissue infrared imaging. Adv Drug Deliv Rev 2013; 65:719-31. [PMID: 22981756 DOI: 10.1016/j.addr.2012.08.016] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 07/11/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022]
Abstract
Near infrared fluorescence offers several advantages for tissue and in vivo imaging thanks to deeper photon penetration. In this article, we review a promising class of near infrared emitting probes based on semiconductor quantum dots (QDs), which have the potential to considerably improve in vivo fluorescence imaging thanks to their high brightness and stability. We discuss in particular the different criteria to optimize the design of near infrared QDs. We present the recent developments in the synthesis of novel QD materials and their different in vivo imaging applications, including lymph node localization, vasculature imaging, tumor localization, as well as cell tracking and QD-based multimodal probes.
Collapse
|
199
|
Probst CE, Zrazhevskiy P, Bagalkot V, Gao X. Quantum dots as a platform for nanoparticle drug delivery vehicle design. Adv Drug Deliv Rev 2013; 65:703-18. [PMID: 23000745 PMCID: PMC3541463 DOI: 10.1016/j.addr.2012.09.036] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 08/19/2012] [Accepted: 09/12/2012] [Indexed: 02/07/2023]
Abstract
Nanoparticle-based drug delivery (NDD) has emerged as a promising approach to improving upon the efficacy of existing drugs and enabling the development of new therapies. Proof-of-concept studies have demonstrated the potential for NDD systems to simultaneously achieve reduced drug toxicity, improved bio-availability, increased circulation times, controlled drug release, and targeting. However, clinical translation of NDD vehicles with the goal of treating particularly challenging diseases, such as cancer, will require a thorough understanding of how nanoparticle properties influence their fate in biological systems, especially in vivo. Consequently, a model system for systematic evaluation of all stages of NDD with high sensitivity, high resolution, and low cost is highly desirable. In theory, this system should maintain the properties and behavior of the original NDD vehicle, while providing mechanisms for monitoring intracellular and systemic nanocarrier distribution, degradation, drug release, and clearance. For such a model system, quantum dots (QDots) offer great potential. QDots feature small size and versatile surface chemistry, allowing their incorporation within virtually any NDD vehicle with minimal effect on overall characteristics, and offer superb optical properties for real-time monitoring of NDD vehicle transport and drug release at both cellular and systemic levels. Though the direct use of QDots for drug delivery remains questionable due to their potential long-term toxicity, the QDot core can be easily replaced with other organic drug carriers or more biocompatible inorganic contrast agents (such as gold and magnetic nanoparticles) by their similar size and surface properties, facilitating translation of well characterized NDD vehicles to the clinic, maintaining NDD imaging capabilities, and potentially providing additional therapeutic functionalities such as photothermal therapy and magneto-transfection. In this review we outline unique features that make QDots an ideal platform for nanocarrier design and discuss how this model has been applied to study NDD vehicle behavior for diverse drug delivery applications.
Collapse
Affiliation(s)
| | | | - Vaishali Bagalkot
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Xiaohu Gao
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
200
|
Geometric control of vascular networks to enhance engineered tissue integration and function. Proc Natl Acad Sci U S A 2013; 110:7586-91. [PMID: 23610423 DOI: 10.1073/pnas.1217796110] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tissue vascularization and integration with host circulation remains a key barrier to the translation of engineered tissues into clinically relevant therapies. Here, we used a microtissue molding approach to demonstrate that constructs containing highly aligned "cords" of endothelial cells triggered the formation of new capillaries along the length of the patterned cords. These vessels became perfused with host blood as early as 3 d post implantation and became progressively more mature through 28 d. Immunohistochemical analysis showed that the neovessels were composed of human and mouse endothelial cells and exhibited a mature phenotype, as indicated by the presence of alpha-smooth muscle actin-positive pericytes. Implantation of cords with a prescribed geometry demonstrated that they provided a template that defined the neovascular architecture in vivo. To explore the utility of this geometric control, we implanted primary rat and human hepatocyte constructs containing randomly organized endothelial networks vs. ordered cords. We found substantially enhanced hepatic survival and function in the constructs containing ordered cords following transplantation in mice. These findings demonstrate the importance of multicellular architecture in tissue integration and function, and our approach provides a unique strategy to engineer vascular architecture.
Collapse
|