351
|
Henshaw JW, Zaharoff DA, Mossop BJ, Yuan F. Electric field-mediated transport of plasmid DNA in tumor interstitium in vivo. Bioelectrochemistry 2007; 71:233-42. [PMID: 17728192 PMCID: PMC2885976 DOI: 10.1016/j.bioelechem.2007.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 07/07/2007] [Accepted: 07/18/2007] [Indexed: 12/11/2022]
Abstract
Local pulsed electric field application is a method for improving non-viral gene delivery. Mechanisms of the improvement include electroporation and electrophoresis. To understand how electrophoresis affects pDNA delivery in vivo, we quantified the magnitude of electric field-induced interstitial transport of pDNA in 4T1 and B16.F10 tumors implanted in mouse dorsal skin-fold chambers. Four different electric pulse sequences were used in this study, each consisted of 10 identical pulses that were 100 or 400 V/cm in strength and 20 or 50 ms in duration. The interval between consecutive pulses was 1 s. The largest distance of transport was obtained with the 400 V/cm and 50 ms pulse, and was 0.23 and 0.22 microm/pulse in 4T1 and B16.F10 tumors, respectively. There were no significant differences in transport distances between 4T1 and B16.F10 tumors. Results from in vivo mapping and numerical simulations revealed an approximately uniform intratumoral electric field that was predominantly in the direction of the applied field. The data in the study suggested that interstitial transport of pDNA induced by a sequence of ten electric pulses was ineffective for macroscopic delivery of genes in tumors. However, the induced transport was more efficient than passive diffusion.
Collapse
Affiliation(s)
| | | | | | - Fan Yuan
- Corresponding author: Dr. Fan Yuan Department of Biomedical Engineering Duke University 136 Hudson Hall Durham, NC 27708 (919) 660 – 5411 (phone) (919) 684 – 4488 (fax)
| |
Collapse
|
352
|
Verma A, Mehta K. Tissue transglutaminase-mediated chemoresistance in cancer cells. Drug Resist Updat 2007; 10:144-51. [PMID: 17662645 DOI: 10.1016/j.drup.2007.06.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 06/08/2007] [Accepted: 06/11/2007] [Indexed: 12/30/2022]
Abstract
Drug resistance and metastasis are major impediments for the successful treatment of cancer. A common feature among drug resistant and metastatic tumor cells is that they exhibit profound resistance to apoptosis. This property enables cancer cells not only to grow and survive in stressful environments (metastasis) but also to display resistance against many anticancer agents. Therefore, perturbation of the intrinsic apoptotic pathways of cancer cells will affect their ability to respond to chemotherapy and to metastasize and survive in distant sites. Recent studies have demonstrated that cancer cells and cancer cell lines selected for resistance against chemotherapeutic drugs or isolated from metastatic sites, express elevated levels of the multifunctional protein, tissue transglutaminase (TG2). TG2 is the most diverse and ubiquitous member of the transglutaminase family of proteins that is implicated to play a role in apoptosis, wound healing, cell migration, cell attachment, cell growth, angiogenesis, and matrix assembly. TG2 can associate with certain beta members of the integrin family of proteins (beta1, beta3, beta4, and beta5) and promote stable interaction between cells and the extracellular matrix (ECM), resulting in increased cell survival, cell migration, and invasion. Additionally, TG2 forms a ternary complex with IkappaB/p65:p50 and results in constitutive activation of the nuclear transcription factor-kappaB (NF-kappaB). Moreover, TG2 expression in cancer cells leads to constitutive activation of the focal adhesion kinase (FAK) and its downstream PI3K/Akt survival pathway. Importantly, the inhibition of endogenous TG2 by small interfering RNA (siRNA) resulted in the reversal of drug resistance and the invasive phenotype. Conversely, ectopic expression of TG2 promoted cell survival, cell motility and invasive functions of cancer cells. This review discusses the current thinking and implications of increased TG2 expression in development of drug resistance and metastasis by cancer cells.
Collapse
Affiliation(s)
- Amit Verma
- Department of Experimental Therapeutics, Unit 362, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States
| | | |
Collapse
|
353
|
Colombo L, González G, Marshall G, Molina FV, Soba A, Suarez C, Turjanski P. Ion transport in tumors under electrochemical treatment: in vivo, in vitro and in silico modeling. Bioelectrochemistry 2007; 71:223-32. [PMID: 17689151 DOI: 10.1016/j.bioelechem.2007.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 06/14/2007] [Accepted: 07/06/2007] [Indexed: 11/24/2022]
Abstract
The electrochemical treatment of cancer (EChT) consists in the passage of a direct electric current through two or more electrodes inserted locally in the tumor tissue. The extreme pH changes induced have been proposed as the main tumor destruction mechanism. Here, we study ion transport during EChT through a combined modeling methodology: in vivo modeling with BALB/c mice bearing a subcutaneous tumor, in vitro modeling with agar and collagen gels, and in silico modeling using the one-dimensional Nernst-Planck and Poisson equations for ion transport in a four-ion electrolyte. This combined modeling approach reveals that, under EChT modeling, an initial condition with almost neutral pH evolves between electrodes into extreme cathodic alkaline and anodic acidic fronts moving towards each other, leaving the possible existence of a biological pH region between them; towards the periphery, the pH decays to its neutral values. pH front tracking unveils a time scaling close to t(1/2), signature of a diffusion-controlled process. These results could have significant implications in EChT optimal operative conditions and dose planning, in particular, in the way in which the evolving EChT pH region covers the active cancer cells spherical casket.
Collapse
Affiliation(s)
- L Colombo
- Depto. de Inmunobiología, Inst. de Oncología Angel H. Roffo, Universidad de Buenos Aires, (C1417DTB) Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
354
|
Wijeratne NS, Hoo KA. Understanding the role of the tumour vasculature in the transport of drugs to solid cancer tumours. Cell Prolif 2007; 40:283-301. [PMID: 17531075 PMCID: PMC6760703 DOI: 10.1111/j.1365-2184.2007.00436.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES The vasculature of tumours imposes certain barriers that transport of anti-cancer drugs must overcome. Here follows an account of development of a general computational model that describes the mechanisms of drug transport to a solid tumour, with an emphasis on modelling the vasculature using solute transport concepts. MATERIALS AND METHODS Investigation into the biological parameters that enhance/prevent anticancer drug transport to the tumour provides a means to evaluate the effects of these parameters on the treatment process. Sensitivity analysis of these provides useful insights concerning anticancer drug transport mechanisms from the vasculature to the solid tumour for a non-specified drug and non-specified solid tumour by revealing the conditions that promote or prevent effective drug transport. The effect of the vasculature on transport efficiency is studied using a parametric analysis of some of the transport and biological parameters. Understanding the various transport mechanisms provides a basis to evaluate the effectiveness of the drug treatment a priori. RESULTS It was found that increases in the capillary hydrostatic pressure, diffusive permeability coefficient and hydraulic conductivity all result in a decrease in tumour size. Similarly, decreases in the interstitium hydrostatic pressure and filtration constant result in a decrease in tumour size. Dependence of the change in the tumour size to changes in these parameters is non-linear. These results demonstrate the potential of the integrated computational model of the tumour and its vasculature to estimate efficacy of a particular treatment process. Regardless of the dependency of the outcome on the assumed model parameters and the assumed kinetics, mathematical models of this type can provide more explanation on the issues related to the transport barriers, the efficacy of the treatment, and the development of effective anticancer drugs. A case study also is presented to demonstrate the model's flexibility to accommodate a two-cell-glioma population.
Collapse
Affiliation(s)
- N S Wijeratne
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | | |
Collapse
|
355
|
Mesojednik S, Pavlin D, Sersa G, Coer A, Kranjc S, Grosel A, Tevz G, Cemazar M. The effect of the histological properties of tumors on transfection efficiency of electrically assisted gene delivery to solid tumors in mice. Gene Ther 2007; 14:1261-9. [PMID: 17597791 DOI: 10.1038/sj.gt.3302989] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Uniform DNA distribution in tumors is a prerequisite step for high transfection efficiency in solid tumors. To improve the transfection efficiency of electrically assisted gene delivery to solid tumors in vivo, we explored how tumor histological properties affected transfection efficiency. In four different tumor types (B16F1, EAT, SA-1 and LPB), proteoglycan and collagen content was morphometrically analyzed, and cell size and cell density were determined in paraffin-embedded tumor sections under a transmission microscope. To demonstrate the influence of the histological properties of solid tumors on electrically assisted gene delivery, the correlation between histological properties and transfection efficiency with regard to the time interval between DNA injection and electroporation was determined. Our data demonstrate that soft tumors with larger spherical cells, low proteoglycan and collagen content, and low cell density are more effectively transfected (B16F1 and EAT) than rigid tumors with high proteoglycan and collagen content, small spindle-shaped cells and high cell density (LPB and SA-1). Furthermore, an optimal time interval for increased transfection exists only in soft tumors, this being in the range of 5-15 min. Therefore, knowledge about the histology of tumors is important in planning electrogene therapy with respect to the time interval between DNA injection and electroporation.
Collapse
Affiliation(s)
- S Mesojednik
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|
356
|
Ganesh S, Gonzalez Edick M, Idamakanti N, Abramova M, Vanroey M, Robinson M, Yun CO, Jooss K. Relaxin-expressing, fiber chimeric oncolytic adenovirus prolongs survival of tumor-bearing mice. Cancer Res 2007; 67:4399-407. [PMID: 17483354 DOI: 10.1158/0008-5472.can-06-4260] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Selective replication of oncolytic viruses in tumor cells provides a promising approach for the treatment of human cancers. One of the limitations observed with oncolytic viruses currently used in the treatment of solid tumors is the inefficient spread of virus throughout the tumor mass following intratumoral injection. Data are presented showing that oncolytic adenoviruses expressing the relaxin gene and containing an Ad5/Ad35 chimeric fiber showed significantly enhanced transduction and increased virus spread throughout the tumor when compared with non-relaxin-expressing, Ad5-based viruses. The increased spread of such viruses throughout tumors correlated well with improved antitumor efficacy and overall survival in two highly metastatic tumor models. Furthermore, nonreplicating viruses expressing relaxin did not increase metastases, suggesting that high level expression of relaxin will not enhance metastatic spread of tumors. In summary, the data show that relaxin may play a role in rearranging matrix components within tumors, which helps recombinant oncolytic adenoviruses to spread effectively throughout the tumor mass and thereby increase the extent of viral replication within the tumor. Expressing relaxin from Ad5/Ad35 fiber chimeric adenoviruses may prove a potent and novel approach to treating patients with cancer.
Collapse
Affiliation(s)
- Shanthi Ganesh
- Cell Genesys, Inc., South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | |
Collapse
|
357
|
Wu J, Lee A, Lu Y, Lee RJ. Vascular targeting of doxorubicin using cationic liposomes. Int J Pharm 2007; 337:329-35. [PMID: 17275230 DOI: 10.1016/j.ijpharm.2007.01.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 12/29/2006] [Accepted: 01/03/2007] [Indexed: 10/23/2022]
Abstract
Tumor vessel has been recognized as an important target for anticancer therapy. Cationic liposomes have been shown to selectively target tumor endothelial cells, thus can potentially be used as a carrier for chemotherapy agents. In this study, cationic liposomes containing 20 mol% cationic lipid dimethyl dioctadecyl ammonium bromide (DDAB) and loaded with doxorubicin (DOX) were prepared and characterized. The cationic liposomal DOX showed 10.8 and 9.1 times greater cytotoxicity than control PEGylated liposomal DOX in KB oral carcinoma and L1210 murine lymphocytic leukemia cells, and 7.7- and 6.8-fold greater cytotoxicity compared to control neutral non-PEGylated liposomal DOX, repectively, in these two cell lines. Although cationic liposomal DOX had higher tumor accumulation at 30 min after intravenous administration compared to control liposomes (p<0.05), DOX uptake of these liposomes at 24h post-injection was similar to that of PEGylated liposomal DOX (p>0.05) and approximately twice the levels of the free drug and non-PEGylated liposomes. In a murine tumor model generated using L1210 cells, increased survival rate was obtained with cationic liposomal DOX treatment compared to free DOX (p<0.01), neutral liposome control (p<0.01), as well as PEGylated liposomes (p<0.05). In conclusion, the cationic liposomal DOX formulation produced superior in vitro cytotoxicity and in vivo antitumor activity, and warrants further investigation.
Collapse
Affiliation(s)
- Jun Wu
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
358
|
Goodman TT, Olive PL, Pun SH. Increased nanoparticle penetration in collagenase-treated multicellular spheroids. Int J Nanomedicine 2007; 2:265-74. [PMID: 17722554 PMCID: PMC2673974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The extracellular matrix of solid tumors presents a transport barrier that restricts nanoparticle penetration, thereby limiting the efficacy of nano-sized delivery vehicles for cancer imaging and therapy. In this study, the effect of nanoparticle size and collagenase treatment on penetration of carboxylated polystyrene nanoparticles was systematically assessed in a multicellular spheroid model. Penetration of the nanoparticles into the spheroid core was limited to particles smaller than 100 nm. Collagenase treatment of spheroids resulted in significantly increased penetration of nanoparticles up to 100 nm with only a minor increase in particle penetration observed for particles larger than 100 nm. Collagenase was immobilized onto the surface of nanoparticles for site-specific degradation of ECM proteins. Collagenase-coated, 100 nm nanoparticles demonstrated a 4-fold increase in the number of particles delivered to the spheroid core compared with control nanoparticles. Thus, nanoparticle delivery to solid tumors may be substantially improved by the incorporation of ECM-modulating enzymes in the delivery formulation.
Collapse
Affiliation(s)
- Thomas T Goodman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Correspondence: Suzie H Pun, University of Washington, Department of Bioengineering, Box 355061, Seattle, WA 98195, USA, Tel +1 206 685 3488, Email
| |
Collapse
|
359
|
Zhou X, Chen B, Hoopes PJ, Hasan T, Pogue BW. Tumor vascular area correlates with photosensitizer uptake: analysis of verteporfin microvascular delivery in the Dunning rat prostate tumor. Photochem Photobiol 2007; 82:1348-57. [PMID: 17421078 DOI: 10.1562/2006-03-25-ra-858] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The parameters that limit supply of photosensitizer to the cancer cells in a solid tumor were systematically analyzed with the use of microvascular transport modeling and histology data from frozen sections. In particular, the vascular permeability transport coefficient and the effective interstitial diffusion coefficient were quantified for Verteporfin-for-Injection delivery of benzoporphyrin derivative (BPD). Orthotopic tumors had higher permeability and diffusion coefficients (Pd = 0.036 microm/s and D = 1.6 microm(2)/s, respectively) as compared to subcutaneously grown tumors (Pd = 0.025 microm/s and D = 0.9 microm2/s, respectively), likely due to the fact that the vessel patterns are more homogeneous orthotopically. In general, large intersubject and intratumor variability exist in the verteporfin concentration, in the range of 25% in plasma concentration and in the range of 20% for tissue concentrations, predominantly due to these microregional variations in transport. However, the average individual uptake of photosensitizer in tumor tissue was only correlated to the total vascular area within the tumor (R2 = 64.1%, P < 0.001). The data are consistent with a view that microregional variation in the vascular permeability and interstitial diffusion rate contribute the spatial heterogeneity observed in verteporfin uptake, but that average supply to the tissue is limited by the total area of perfused blood vessels. This study presents a method to systematically analyze micro-heterogeneity as well as possible methods to increase delivery and homogeneity of photosensitizer within tumor tissue.
Collapse
Affiliation(s)
- Xiaodong Zhou
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | | | | | | | | |
Collapse
|
360
|
Zhou X, Pogue BW, Chen B, Hasan T. Analysis of Effective Molecular Diffusion Rates for Verteporfin in Subcutaneous Versus Orthotopic Dunning Prostate Tumors ¶. Photochem Photobiol 2007. [DOI: 10.1111/j.1751-1097.2004.tb00016.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
361
|
Braeckmans K, Remaut K, Vandenbroucke RE, Lucas B, De Smedt SC, Demeester J. Line FRAP with the confocal laser scanning microscope for diffusion measurements in small regions of 3-D samples. Biophys J 2007; 92:2172-83. [PMID: 17208970 PMCID: PMC1861797 DOI: 10.1529/biophysj.106.099838] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2006] [Accepted: 11/28/2006] [Indexed: 11/18/2022] Open
Abstract
We present a truly quantitative fluorescence recovery after photobleaching (FRAP) model for use with the confocal laser scanning microscope based on the photobleaching of a long line segment. The line FRAP method is developed to complement the disk FRAP method reported before. Although being more subject to the influence of noise, the line FRAP model has the advantage of a smaller bleach region, thus allowing for faster and more localized measurements of the diffusion coefficient and mobile fraction. The line FRAP model is also very well suited to examine directly the influence of the bleaching power on the effective bleaching resolution. We present the outline of the mathematical derivation, leading to a final analytical expression to calculate the fluorescence recovery. We examine the influence of the confocal aperture and the bleaching power on the measured diffusion coefficient to find the optimal experimental conditions for the line FRAP method. This will be done for R-phycoerythrin and FITC-dextrans of various molecular weights. The ability of the line FRAP method to measure correctly absolute diffusion coefficients in three-dimensional samples will be evaluated as well. Finally we show the application of the method to the simultaneous measurement of free green fluorescent protein diffusion in the cytoplasm and nucleus of living A549 cells.
Collapse
Affiliation(s)
- Kevin Braeckmans
- Laboratory General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | | | | | | | | | | |
Collapse
|
362
|
Jain M, Venkatraman G, Batra SK. Optimization of radioimmunotherapy of solid tumors: biological impediments and their modulation. Clin Cancer Res 2007; 13:1374-82. [PMID: 17309914 DOI: 10.1158/1078-0432.ccr-06-2436] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In contrast to the overwhelming success of radiolabeled antibodies in treating hematologic malignancies, only modest success has been achieved in the radioimmunotherapy of solid tumors. One of the major limitations in successful application of radioimmunotherapy is the large molecular size of the intact immunoglobulin that results in prolonged serum half-life and poor tumor penetration and uptake. With the advent of antibody engineering, small molecular weight antibody fragments exhibiting improved pharmacokinetics and tumor penetration have been generated. However, their clinical application has been limited by suboptimal tumor uptake and short tumor residence time. There is a greater realization that optimization of the molecular size of the antibodies alone is not sufficient for clinical success of radioimmunotherapy. In addition to their size, radiolabeled antibodies encounter other impediments before reaching their target antigens expressed on the cell surface of solid tumors. Some of the barriers include poor blood flow in large tumors, permeability of vascular endothelium, elevated interstitial fluid pressure of tumor stroma, and heterogeneous antigen expression. Recent research has considerably improved our understanding and appreciation of these forces, and the new wave of optimization strategies involves the use of biological modifiers to modulate the impediments posed by solid tumors. In combination with radiolabeled antibodies, various agents are being used to improve the tumor blood flow, enhance vascular permeability, lower tumor interstitial fluid pressure by modulating stromal cells and extracellular matrix components, up-regulate the expression of target antigens, and improve the penetration and retention of the radiopharmaceuticals. This review outlines ongoing research efforts involving biological modifiers to optimize the uptake and efficacy of radiolabeled antibodies for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maneesh Jain
- Department of Biochemistry and Molecular Biology, Department of Pathology and Microbiology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | | | | |
Collapse
|
363
|
Abstract
Whereas over 85% of human cancers are solid tumors, of the 8 monoclonal antibodies (mAbs) currently approved for cancer therapy, 25% are directed at solid tumor surface antigens (Ags). This shortfall may be due to barriers to achieving adequate exposure in solid tumors. Advancements in tumor biology, protein engineering, and theoretical modeling of macromolecular transport are currently enabling identification of critical physical properties for antitumor Abs. It is now possible to structurally modify Abs or even replace full Abs with a plethora of Ab constructs. These constructs include Fab and Fab'(2) fragments, scFvs, multivalent scFvs (e.g., diabodies and tribodies), minibodies (e.g., scFv-CH3 dimers), bispecific Abs, and camel variable functional heavy chain domains. The purpose of the article is to provide investigators with a conceptual framework for exploiting the recent scientific advancements. The focus is on 2 properties that govern tumor exposure: 1) physical properties that enable penetration of and retention by tumors, and 2) favorable plasma pharmacokinetics. It is demonstrated that manipulating molecular size, charge, valence, and binding affinity can optimize these properties. These manipulations hold the key to promoting tumor exposure and to ultimately creating successful Ab therapies for solid tumors.
Collapse
Affiliation(s)
- Robert A Beckman
- Clinical Hematology-Oncology, Centocor Research and Development, Inc., Malvern, Pennsylvania 19355, USA.
| | | | | |
Collapse
|
364
|
Hickey JD, Heller L, Heller R, Gilbert R. Electric field mediated DNA motion model. Bioelectrochemistry 2007; 70:101-3. [PMID: 16714149 DOI: 10.1016/j.bioelechem.2006.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Indexed: 11/20/2022]
Abstract
Understanding the motion and the governing equations of a molecule's path in tissue is an ultimate requirement for the repeatable, site specific delivery of molecules [Joseph D. Hickey. Modelling the Motion of Ions and Molecules in Electroporation and Electrophoresis Field Conditions. University of South Florida, College of Arts and Sciences, Department of Physics, Tampa, Florida, 2003., Joseph D. Hickey and Richard Gilbert. Modeling the electromobility of ions in a target tissue. DNA and Cell Biology, 22 (12) (2003) 823-828.]. This paper describes a computationally efficient mathematical model and simulation technique for the examination of DNA fragments in a 1% agarose gel. The speed of the individual DNA fragments through the agarose gel was described through two parts. The maximum velocity was calculated using the Coulombic force divided by Stoke's law and that value was retarded by an exponential rate equation. The simulation utilizes previously published techniques modified for this specific application [Joseph D. Hickey and Richard Gilbert. Fluid flow electrophoresis model. Bioelectrochemistry, 63 (2) (2004) 365-367., Joseph D. Hickey and Richard Gilbert. Modeling the electromobility of ions in a target tissue. DNA and Cell Biology, 22 (12) (2003) 823-828.]. Five representative DNA fragment sizes that span the resolution of a 1% agarose gel were chosen for this analysis. The speeds corresponding to these five DNA fragment sizes were converted into discrete values and used in a 50 step simulation. The resultant error comparing the simulation with experimental distance was 7.76%. Through a 1-D optimization procedure, this error was reduced to 3.02% for a 52 step simulation.
Collapse
Affiliation(s)
- Joseph D Hickey
- Department of Chemical Engineering, University of South Florida, 4202 E. Fowler Ave ENB 118, Tampa, Fl 33620, USA.
| | | | | | | |
Collapse
|
365
|
Henshaw JW, Zaharoff DA, Mossop BJ, Yuan F. A single molecule detection method for understanding mechanisms of electric field-mediated interstitial transport of genes. Bioelectrochemistry 2006; 69:248-53. [PMID: 16713747 DOI: 10.1016/j.bioelechem.2006.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 03/20/2006] [Accepted: 03/22/2006] [Indexed: 10/24/2022]
Abstract
The interstitial space is a rate limiting physiological barrier to non-viral gene delivery. External pulsed electric fields have been proposed to increase DNA transport in the interstitium, thereby improving non-viral gene delivery. In order to characterize and improve the interstitial transport, we developed a reproducible single molecule detection method to observe the electromobility of DNA in a range of pulsed, high field strength electric fields typically used during electric field-mediated gene delivery. Using agarose gel as an interstitium phantom, we investigated the dependence of DNA electromobility on field magnitude, pulse duration, pulse interval, and pore size in the interstitial space. We observed that the characteristic electromobility behavior, exhibited under most pulsing conditions, consisted of three distinct phases: stretching, reptation, and relaxation. Electromobility depended strongly on the field magnitude, pulse duration, and pulse interval of the applied pulse sequences, as well as the pore size of the fibrous matrix through which the DNA migrated. Our data also suggest the existence of a minimum pulse amplitude required to initiate electrophoretic transport. These results are useful for understanding the mechanisms of DNA electromobility and improving interstitial transport of genes during electric field-mediated gene delivery.
Collapse
Affiliation(s)
- Joshua W Henshaw
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
366
|
Cabrera G, Porvasnik SL, DiCorleto PE, Siemionow M, Goldman CK. Intra-arterial adenoviral mediated tumor transfection in a novel model of cancer gene therapy. Mol Cancer 2006; 5:32. [PMID: 16899125 PMCID: PMC1560393 DOI: 10.1186/1476-4598-5-32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 08/09/2006] [Indexed: 11/26/2022] Open
Abstract
Background The aim of the present study was to develop and characterize a novel in vivo cancer gene therapy model in which intra-arterial adenoviral gene delivery can be characterized. In this model, the rat cremaster muscle serves as the site for tumor growth and provides convenient and isolated access to the tumor parenchyma with discrete control of arterial and venous access for delivery of agents. Results Utilizing adenovirus encoding the green fluorescent protein we demonstrated broad tumor transfection. We also observed a dose dependant increment in luciferase activity at the tumor site using an adenovirus encoding the luciferase reporter gene. Finally, we tested the intra-arterial adenovirus dwelling time required to achieve optimal tumor transfection and observed a minimum time of 30 minutes. Conclusion We conclude that adenovirus mediated tumor transfection grown in the cremaster muscle of athymic nude rats via an intra-arterial route could be achieved. This model allows definition of the variables that affect intra-arterial tumor transfection. This particular study suggests that allowing a defined intra-tumor dwelling time by controlling the blood flow of the affected organ during vector infusion can optimize intra-arterial adenoviral delivery.
Collapse
Affiliation(s)
- Gustavo Cabrera
- Gene Therapy Laboratory, National Cancer Institute, Mexico City, Mexico
| | - Stacy L Porvasnik
- Powel Gene Therapy Center, The University of Florida, Gainesville, USA
| | - Paul E DiCorleto
- Department of Cell Biology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, USA
| | - Maria Siemionow
- Department of Plastic and Reconstructive Surgery, The Cleveland Clinic Foundation, Cleveland, USA
| | - Corey K Goldman
- Department of Vascular Medicine, Ochsner Clinic Foundation, New Orleans, USA
| |
Collapse
|
367
|
Ludwig JA, Szakács G, Martin SE, Chu BF, Cardarelli C, Sauna ZE, Caplen NJ, Fales HM, Ambudkar SV, Weinstein JN, Gottesman MM. Selective toxicity of NSC73306 in MDR1-positive cells as a new strategy to circumvent multidrug resistance in cancer. Cancer Res 2006; 66:4808-15. [PMID: 16651436 PMCID: PMC1474781 DOI: 10.1158/0008-5472.can-05-3322] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ATP-binding cassette (ABC) proteins include the best known mediators of resistance to anticancer drugs. In particular, ABCB1 [MDR1/P-glycoprotein (P-gp)] extrudes many types of drugs from cancer cells, thereby conferring resistance to those agents. Attempts to overcome P-gp-mediated drug resistance using specific inhibitors of P-gp has had limited success and has faced many therapeutic challenges. As an alternative approach to using P-gp inhibitors, we characterize a thiosemicarbazone derivative (NSC73306) identified in a generic screen as a compound that exploits, rather than suppresses, P-gp function to induce cytotoxicity. Cytotoxic activity of NSC73306 was evaluated in vitro using human epidermoid, ovarian, and colon cancer cell lines expressing various levels of P-gp. Our findings suggest that cells become hypersensitive to NSC73306 in proportion to the increased P-gp function and multidrug resistance (MDR). Abrogation of both sensitivity to NSC73306 and resistance to P-gp substrate anticancer agents occurred with specific inhibition of P-gp function using either a P-gp inhibitor (PSC833, XR9576) or RNA interference, suggesting that cytotoxicity was linked to MDR1 function, not to other, nonspecific factors arising during the generation of resistant or transfected cells. Molecular characterization of cells selected for resistance to NSC73306 revealed loss of P-gp expression and consequent loss of the MDR phenotype. Although hypersensitivity to NSC73306 required functional expression of P-gp, biochemical assays revealed no direct interaction between NSC73306 and P-gp. This article shows that NSC73306 kills cells with intrinsic or acquired P-gp-induced MDR and indirectly acts to eliminate resistance to MDR1 substrates.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/metabolism
- Cyclosporins/pharmacology
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor
- Drug Synergism
- Female
- Humans
- Indoles/pharmacology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- RNA, Small Interfering/genetics
Collapse
Affiliation(s)
- Joseph A. Ludwig
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
- Genomics and Bioinformatics Group, Laboratory of Molecular Pharmacology, CCR, NCI, NIH
| | - Gergely Szakács
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| | - Scott E. Martin
- Gene Silencing Section, Office of Science and Technology Partnerships, Office of the Director, CCR, NCI, NIH
| | - Benjamin F. Chu
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| | - Carol Cardarelli
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| | - Zuben E. Sauna
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| | - Natasha J. Caplen
- Gene Silencing Section, Office of Science and Technology Partnerships, Office of the Director, CCR, NCI, NIH
| | - Henry M. Fales
- Laboratory of Biophysical Chemistry, National Heart Lung and Blood Institute, NIH
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| | - John N. Weinstein
- Genomics and Bioinformatics Group, Laboratory of Molecular Pharmacology, CCR, NCI, NIH
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland 20892
| |
Collapse
|
368
|
Thiagarajah JR, Kim JK, Magzoub M, Verkman AS. Slowed diffusion in tumors revealed by microfiberoptic epifluorescence photobleaching. Nat Methods 2006; 3:275-80. [PMID: 16554832 DOI: 10.1038/nmeth863] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 02/09/2006] [Indexed: 11/09/2022]
Abstract
It has not been possible to measure diffusion deep in solid tissues such as tumors because of the limited light penetration of conventional optical techniques. Here we report a microfiberoptic epifluorescence photobleaching (MFEP) method in which photobleaching is done by laser epi-illumination through a multimode fiberoptic whose micron-sized tip can be introduced deep into tissues. We applied MFEP to measure the diffusion of fluorescent macromolecules in tumors in living mice, at depths well beyond those accessible by surface optical measurements. Macromolecule diffusion was slowed about twofold within 200 microm of the surface of a solid tumor, but was slowed greater than tenfold beyond 500 microm. Our results reveal a remarkable and previously unrecognized slowing of diffusion deep in tumors, which correlated with the differing tissue architectures of tumor periphery versus core, and with altered tumor vasculature produced by aquaporin-1 deletion. MFEP should have wide applications for measuring diffusion in organs, solid tumors and other light-inaccessible tissue masses.
Collapse
Affiliation(s)
- Jay R Thiagarajah
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
369
|
Siepmann J, Siepmann F, Florence AT. Local controlled drug delivery to the brain: mathematical modeling of the underlying mass transport mechanisms. Int J Pharm 2006; 314:101-19. [PMID: 16647231 DOI: 10.1016/j.ijpharm.2005.07.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 07/12/2005] [Indexed: 10/24/2022]
Abstract
The mass transport mechanisms involved in the controlled delivery of drugs to living brain tissue are complex and yet not fully understood. Often the drug is embedded within a polymeric or lipidic matrix, which is directly administered into the brain tissue, that is, intracranially. Different types of systems, including microparticles and disc- or rod-shaped implants are used to control the release rate and, thus, to optimize the drug concentrations at the site of action in the brain over prolonged periods of time. Most of these dosage forms are biodegradable to avoid the need for the removal of empty remnants after drug exhaustion. Various physical and chemical processes are involved in the control of drug release from these systems, including water penetration, drug dissolution, degradation of the matrix and drug diffusion. Once the drug has been released from the delivery system, it has to be transported through the living brain tissue to the target site(s). Again, a variety of phenomena, including diffusion, drug metabolism and degradation, passive or active uptake into CNS tissue and convection can be of importance for the fate of the drug. An overview is given of the current knowledge of the nature of barriers to free access of drug to tumour sites within the brain and the state of the art of: (i) mathematical modeling approaches describing the physical transport processes and chemical reactions which can occur in different types of intracranially administered drug delivery systems, and of (ii) theories quantifying the mass transport phenomena occurring after drug release in the living tissue. Both, simplified as well as complex mathematical models are presented and their major advantages and shortcomings discussed. Interestingly, there is a significant lack of mechanistically realistic, comprehensive theories describing both parts in detail, namely, drug transport in the dosage form and in the living brain tissue. High quality experimental data on drug concentrations in the brain tissue are difficult to obtain, hence this is itself an issue in testing mathematical approaches. As a future perspective, the potential benefits and limitations of these mathematical theories aiming to facilitate the design of advanced intracranial drug delivery systems and to improve the efficiency of the respective pharmacotherapies are discussed.
Collapse
Affiliation(s)
- J Siepmann
- College of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany.
| | | | | |
Collapse
|
370
|
Ky D, Liu CK, Marumoto C, Castaneda L, Slowinska K. Electrochemical Time-of-Flight in crosslinked collagen matrix solution: Implications of structural changes for drug delivery systems. J Control Release 2006; 112:214-22. [PMID: 16564597 DOI: 10.1016/j.jconrel.2006.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Revised: 02/01/2006] [Accepted: 02/08/2006] [Indexed: 11/25/2022]
Abstract
Electrochemical Time-of-Flight (ETOF) method was used for the first time to measure the diffusion coefficient of 4-hydroxy-TEMPO, a molecular probe in collagen I matrix solution as a function of its concentration and the extent of crosslinking with glutaraldehyde (GA). The values of the diffusion coefficient were correlated with Circular Dichroism (CD) and viscosity data to assess the changes of the structure of a collagen matrix. The low value of probe diffusion coefficient indicates that the molecular collagen contributes to large diffusion hindrance of the medium. The combined Brinkman or effective medium model and the Carman-Kozeny model were used to estimate the average diameter of a matrix pore as a function of collagen solution composition. We show that 0.5% and 1% (w/w) collagen matrix crosslinked with the addition of GA above 0.1% (v/w) forms matrix with pores larger than in native collagen. This result suggests the existence of a micro-phase separation in the crosslinked collagen matrix. The implications of these findings for the design of small molecule drug delivery systems are discussed.
Collapse
Affiliation(s)
- Darline Ky
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, CA 90840, USA
| | | | | | | | | |
Collapse
|
371
|
McKee TD, Grandi P, Mok W, Alexandrakis G, Insin N, Zimmer JP, Bawendi MG, Boucher Y, Breakefield XO, Jain RK. Degradation of fibrillar collagen in a human melanoma xenograft improves the efficacy of an oncolytic herpes simplex virus vector. Cancer Res 2006; 66:2509-13. [PMID: 16510565 DOI: 10.1158/0008-5472.can-05-2242] [Citation(s) in RCA: 320] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oncolytic viral therapy provides a promising approach to treat certain human malignancies. These vectors improve on replication-deficient vectors by increasing the viral load within tumors through preferential viral replication within tumor cells. However, the inability to efficiently propagate throughout the entire tumor and infect cells distant from the injection site has limited the capacity of oncolytic viruses to achieve consistent therapeutic responses. Here we show that the spread of the oncolytic herpes simplex virus (HSV) vector MGH2 within the human melanoma Mu89 is limited by the fibrillar collagen in the extracellular matrix. This limitation seems to be size specific as nanoparticles of equivalent size to the virus distribute within tumors to the same extent whereas smaller particles distribute more widely. Due to limited viral penetration, tumor cells in inaccessible regions continue to grow, remaining out of the range of viral infection, and tumor eradication cannot be achieved. Matrix modification with bacterial collagenase coinjection results in a significant improvement in the initial range of viral distribution within the tumor. This results in an extended range of infected tumor cells and improved virus propagation, ultimately leading to enhanced therapeutic outcome. Thus, fibrillar collagen can be a formidable barrier to viral distribution and matrix-modifying treatments can significantly enhance the therapeutic response.
Collapse
Affiliation(s)
- Trevor D McKee
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachussetts 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
372
|
Nakahara T, Norberg SM, Shalinsky DR, Hu-Lowe DD, McDonald DM. Effect of inhibition of vascular endothelial growth factor signaling on distribution of extravasated antibodies in tumors. Cancer Res 2006; 66:1434-45. [PMID: 16452199 DOI: 10.1158/0008-5472.can-05-0923] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibodies and other macromolecular therapeutics can gain access to tumor cells via leaky tumor vessels. Inhibition of vascular endothelial growth factor (VEGF) signaling can reduce the vascularity of tumors and leakiness of surviving vessels, but little is known about how these changes affect the distribution of antibodies within tumors. We addressed this issue by examining the distribution of extravasated antibodies in islet cell tumors of RIP-Tag2 transgenic mice and implanted Lewis lung carcinomas using fluorescence and confocal microscopic imaging. Extravasated nonspecific immunoglobulin G (IgG) and antibodies to fibrin or E-cadherin accumulated in irregular patchy regions of stroma. Fibrin also accumulated in these regions. Anti-E-cadherin antibody, which targets epitopes on tumor cells of RIP-Tag2 adenomas, was the only antibody to achieve detectable levels within tumor cell clusters at 6 hours after i.v. injection. Treatment for 7 days with AG-013736, a potent inhibitor of VEGF signaling, reduced the tumor vascularity by 86%. The overall area density of extravasated IgG/antibodies decreased after treatment but the change was less than the reduction in vascularity and actually increased when expressed per surviving tumor vessel. Accumulation of anti-E-cadherin antibody in tumor cell clusters was similarly affected. The patchy pattern of antibodies in stroma after treatment qualitatively resembled untreated tumors and surprisingly coincided with sleeves of basement membrane left behind after pruning of tumor vessels. Together, the findings suggest that antibody transport increases from surviving tumor vessels after normalization by inhibition of VEGF signaling. Basement membrane sleeves may facilitate this transport. Antibodies preferentially distribute to tumor stroma but also accumulate on tumor cells if binding sites are accessible.
Collapse
MESH Headings
- Adenoma, Islet Cell/blood
- Adenoma, Islet Cell/blood supply
- Adenoma, Islet Cell/immunology
- Animals
- Antibodies, Neoplasm/blood
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/metabolism
- Axitinib
- Cadherins/immunology
- Carcinoma, Lewis Lung/blood
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/immunology
- Fibrin/immunology
- Fibrin/metabolism
- Imidazoles/pharmacology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Indazoles/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microspheres
- Neovascularization, Pathologic/blood
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Signal Transduction
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Tsutomu Nakahara
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0130, USA
| | | | | | | | | |
Collapse
|
373
|
Dreher MR, Liu W, Michelich CR, Dewhirst MW, Yuan F, Chilkoti A. Tumor vascular permeability, accumulation, and penetration of macromolecular drug carriers. J Natl Cancer Inst 2006; 98:335-44. [PMID: 16507830 DOI: 10.1093/jnci/djj070] [Citation(s) in RCA: 665] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Delivery of anticancer therapeutic agents to solid tumors is problematic. Macromolecular drug carriers are an attractive alternative drug delivery method because they appear to target tumors and have limited toxicity in normal tissues. We investigated how molecular weight influences the accumulation of a model macromolecular drug carrier, dextran covalently linked to a fluorophore, in tumors. METHODS We used dextrans with molecular weights from 3.3 kDa to 2 MDa. Vascular permeability, accumulation, and three-dimensional penetration of these dextrans were simultaneously measured in solid tumors via a dorsal skin fold window chamber, intravital laser-scanning confocal microscopy, and custom image analysis. RESULTS Increasing the molecular weight of dextran statistically significantly reduced its vascular permeability by approximately two orders of magnitude (i.e., from 154 x 10(-7) cm/s, 95% confidence interval [CI] = 134 to 174 x 10(-7) cm/s, for 3.3-kDa dextran to 1.7 x 10(-7) cm/s, 95% CI = 0.7 to 2.6 x 10(-7) cm/s for 2-MDa dextran; P < .001, two-sided Kruskal-Wallis test) but increased its plasma half-life, which provided ample time for extravasation (i.e., to enter tumor tissue from the vasculature). Tumor accumulation was maximal for dextrans with molecular weights between 40 and 70 kDa. Dextrans of 3.3 and 10 kDa penetrated deeply (greater than 35 microm) and homogeneously into tumor tissue from the vessel wall. After a 30-minute period, a high concentration was observed only approximately 15 microm from the vessel wall for 40- to 70-kDa dextrans and only 5 microm for 2-MDa dextrans. CONCLUSIONS Increasing the molecular weight of dextran statistically significantly reduced its tumor vascular permeability. Dextrans of 40 and 70 kDa had the highest accumulation in solid tumors but were largely concentrated near the vascular surface.
Collapse
Affiliation(s)
- Matthew R Dreher
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
374
|
Kuhn SJ, Hallahan DE, Giorgio TD. Characterization of Superparamagnetic Nanoparticle Interactions with Extracellular Matrix in an in Vitro System. Ann Biomed Eng 2006; 34:51-8. [PMID: 16477503 DOI: 10.1007/s10439-005-9004-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Accepted: 08/30/2005] [Indexed: 11/26/2022]
Abstract
Controlled dispersion of therapeutic agents within liquid- and gel-filled cavities represents a barrier to treatment of some cancers and other pathological states. Interstitial delivery is compromised by the poor mobility of macromolecules and larger nanoscale structures. We developed an in vitro system to quantify the suitability of superparamagnetic nanoparticles (SPM NPs) as a site-specific therapeutic vehicle for delivery through fluid- and gel-based systems. SPM NP motion was induced by an external magnetic field. NP migration was modulated by NP concentration and surface coating. 135 nanometer radius PEGylated NPs moved through the extracellular matrix with an average velocity of 1.5 mm h(-1), suitable for some clinical applications. Increasing the SPM NP radius to 400 nm while maintaining the same per NP magnetic susceptibility resulted in a greater than 1,000-fold reduction in magnetic mobility, to less than 0.01 mm h(-1). The critical influence of NP size on gel permeation was also observed in silica-coated 135 nm SPM NPs that aggregated under the experimental conditions. Aggregation played a critical role in determining the behavior of the nanoparticles. SPM NPs allow significant free-solution mobility to specific sites within a cavity and generate sufficient force to penetrate common in vivo gels.
Collapse
Affiliation(s)
- Sam J Kuhn
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232-8180, USA
| | | | | |
Collapse
|
375
|
Paciotti GF, Kingston DG, Tamarkin L. Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectors. Drug Dev Res 2006. [DOI: 10.1002/ddr.20066] [Citation(s) in RCA: 345] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
376
|
Tatarkova SA, Kamra Verma A, Berk DA, Lloyd CJ. Quantitative fluorescence microscopy of macromolecules in gel and biological tissue. Phys Med Biol 2005; 50:5759-68. [PMID: 16306666 DOI: 10.1088/0031-9155/50/23/024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Quantitative fluorescence microscopy provides valuable insight into drug delivery and pharmacokinetics. The technique is based on analysis of statistical fluctuations in fluorescence that arises as fluorophores pass through a small volume illuminated by a focused laser beam, and has been applied to measure particle motion and binding interactions in solutions, on surfaces and inside the cells. We examined the use of fluorescence correlation spectroscopy combined with a microscope (FCSM) to assess the transport of fluorescent beads and macromolecules in aqueous solutions, gels and living biological tissue. Obstructed diffusion of fluorescent beads in gels of various densities was tested to get a sensible estimate of diffusion in the interstitial tissue matrix consistent with previous reports. Fluorescently labelled liposomes as an artificial drug or gene carrying vehicles were used for pharmacokinetic tests of drug delivery in living tissue. The results indicate that FCS is an accurate and valuable tool for measuring the physical properties of gene vectors in vitro and for characterizing interactions with tissue in vivo.
Collapse
|
377
|
Baranowska-Kortylewicz J, Abe M, Pietras K, Kortylewicz ZP, Kurizaki T, Nearman J, Paulsson J, Mosley RL, Enke CA, Ostman A. Effect of platelet-derived growth factor receptor-beta inhibition with STI571 on radioimmunotherapy. Cancer Res 2005; 65:7824-31. [PMID: 16140951 PMCID: PMC1363769 DOI: 10.1158/0008-5472.can-04-3991] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Whereas radioimmunotherapy of hematologic malignancies has evolved into a viable treatment option, the responses of solid tumors to radioimmunotherapy are discouraging. The likely cause of this problem is the interstitial hypertension inherent to all solid tumors. Remarkable improvements in tumor responses to radioimmunotherapy were discovered after the inclusion of STI571 in the therapy regimen. A combination of the tumor stroma-reactive STI571, a potent platelet-derived growth factor receptor-beta (PDGFr-beta) antagonist, and the tumor-seeking radiolabeled antibody B72.3 yielded long-lasting growth arrest of the human colorectal adenocarcinoma LS174T grown as s.c. xenografts in athymic mice. The interaction of STI571 with the stromal PDGFr-beta reduced tumor interstitial fluid pressure (P(IF)) by >50% and in so doing improved the uptake of B72.3. The attenuation of P(IF) also had a positive effect on the homogeneity of antibody distribution. These effects were dose-dependent and under optimized dosing conditions allowed for a 2.45 times increase in the tumor uptake of B72.3 as determined in the biodistribution studies. Single-photon emission computed tomography imaging studies substantiated these results and indicated that the homogeneity of the radioisotope distribution was also much improved when compared with the control mice. The increased uptake of radioimmunotherapy into the tumor resulted in >400% increase in the tumor absorbed radiation doses in STI571 + radioimmunotherapy-treated mice compared with PBS + radioimmunotherapy-treated mice. The improved antibody uptake in response to the attenuation of tumor P(IF) was identified as the primary reason for the growth arrest of the STI571 + radioimmunotherapy-treated tumors. Two related causes were also identified: (a) the improved homogeneity of monoclonal antibody distribution in tumor and (b) the increased tumor radiosensitivity resulting from the improved tumor oxygenation.
Collapse
|
378
|
Abstract
Applications of nanotechnology for treatment, diagnosis, monitoring, and control of biological systems has recently been referred to as "nanomedicine" by the National Institutes of Health. Research into the rational delivery and targeting of pharmaceutical, therapeutic, and diagnostic agents is at the forefront of projects in nanomedicine. These involve the identification of precise targets (cells and receptors) related to specific clinical conditions and choice of the appropriate nanocarriers to achieve the required responses while minimizing the side effects. Mononuclear phagocytes, dendritic cells, endothelial cells, and cancers (tumor cells, as well as tumor neovasculature) are key targets. Today, nanotechnology and nanoscience approaches to particle design and formulation are beginning to expand the market for many drugs and are forming the basis for a highly profitable niche within the industry, but some predicted benefits are hyped. This article will highlight rational approaches in design and surface engineering of nanoscale vehicles and entities for site-specific drug delivery and medical imaging after parenteral administration. Potential pitfalls or side effects associated with nanoparticles are also discussed.
Collapse
Affiliation(s)
- S Moein Moghimi
- Molecular Targeting and Polymer Toxicology Group, School of Pharmacy, University of Brighton, Brighton, UK
| | | | | |
Collapse
|
379
|
Eikenes L, Tari M, Tufto I, Bruland OS, de Lange Davies C. Hyaluronidase induces a transcapillary pressure gradient and improves the distribution and uptake of liposomal doxorubicin (Caelyx) in human osteosarcoma xenografts. Br J Cancer 2005; 93:81-8. [PMID: 15942637 PMCID: PMC2361478 DOI: 10.1038/sj.bjc.6602626] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Liposomal drug delivery enhances the tumour selective localisation and may improve the uptake compared to free drug. However, the drug distribution within the tumour tissue may still be heterogeneous. Degradation of the extracellular matrix is assumed to improve the uptake and penetration of drugs. The effect of the ECM-degrading enzyme hyaluronidase on interstitial fluid pressure and microvascular pressure were measured in human osteosarcoma xenografts by the wick-in-needle and micropipette technique, respectively. The tumour uptake and distribution of liposomal doxorubicin were studied on tumour sections by confocal laser scanning microscopy. The drugs were injected i.v. 1 h after the hyaluronidase pretreatment. Intratumoral injection of hyaluronidase reduced interstitial fluid pressure in a nonlinear dose-dependent manner. Maximum interstitial fluid pressure reduction of approximately 50% was found after injection of 1500 U hyaluronidase. Neither intratumoral nor i.v. injection of hyaluronidase induced any changes in the microvascular pressure. Thus, hyaluronidase induced a transcapillary pressure gradient, resulting in a four-fold increase in the tumour uptake and improving the distribution of the liposomal doxorubicin. Hyaluronidase reduces a major barrier for drug delivery by inducing a transcapillary pressure gradient, and administration of hyaluronidase adjuvant with liposomal doxorubicin may thus improve the therapeutic outcome.
Collapse
Affiliation(s)
- L Eikenes
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway.
| | | | | | | | | |
Collapse
|
380
|
Papadopoulos MC, Kim JK, Verkman AS. Extracellular space diffusion in central nervous system: anisotropic diffusion measured by elliptical surface photobleaching. Biophys J 2005; 89:3660-8. [PMID: 16143636 PMCID: PMC1366858 DOI: 10.1529/biophysj.105.068114] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Diffusion in the extracellular space (ECS) is crucial for normal central nervous system physiology. The determinants of ECS diffusion include viscous interactions with extracellular matrix/plasma membranes ("viscosity") and ECS geometry ("tortuosity"). To resolve viscosity versus tortuosity effects, we measured direction-dependent (anisotropic) diffusion in ECS in mouse spinal cord by photobleaching using an elliptical spot produced by a cylindrical lens in the excitation path. Anisotropic diffusion slowed fluorescence recovery when the long axis of the ellipse was parallel versus perpendicular to the direction of faster diffusion. A mathematical model was constructed to deduce diffusion coefficients (D(x), D(y)) from fluorescence recovery measured for parallel and perpendicular orientations of the long axis of the ellipse. Elliptical spot photobleaching was validated by photobleaching aqueous-phase fluorophores on a diffraction grating, where diffusion is one-dimensional. Measurement of the diffusion of 70 kDa FITC-dextran in spinal cord in living mice indicated that viscosity slows diffusion by approximately 1.8-fold compared with its diffusion in solution. ECS geometry hinders diffusion across (but not along) axonal fibers in spinal cord further by approximately fivefold. In cerebral cortex, however, approximately 50% of the hindrance to ECS diffusion comes from viscosity and approximately 50% from tortuosity. We suggest that the extracellular matrix might have evolved to facilitate rather than hinder diffusion even for large molecules.
Collapse
Affiliation(s)
- Marios C Papadopoulos
- Departments of Medicine and Physiology, Cardiovascular Research Institute, 1246 Health Sciences East Tower, University of California-San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
381
|
Flessner MF, Choi J, Credit K, Deverkadra R, Henderson K. Resistance of tumor interstitial pressure to the penetration of intraperitoneally delivered antibodies into metastatic ovarian tumors. Clin Cancer Res 2005; 11:3117-25. [PMID: 15837768 DOI: 10.1158/1078-0432.ccr-04-2332] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite evidence that regional chemotherapy improves the treatment of metastatic peritoneal ovarian carcinoma, monoclonal antibodies have not shown significant success in i.p. delivery. The present study was designed to address the hypothesis that convective penetration of macromolecular antineoplastic agents depends on a positive pressure difference between the i.p. therapeutic solution and the tumor. EXPERIMENTAL DESIGN Nude rats with human ovarian xenografts implanted in the abdominal wall were used in experiments to facilitate in vivo measurement of tumor pressure and the treatment of the tumor with i.p. solutions at high pressures. Penetration of (125)I-labeled trastuzumab was measured with quantitative autoradiography. RESULTS Tumor pressure profiles showed peak pressures of 32 mm Hg with mean pressures (+/- SD, mm Hg) in 12 SKOV3 tumors of 9.7 +/- 8.3 and in 15 OVCAR3 tumors of 12.5 +/- 7.0. I.p. therapeutic dwells at 6 to 8 mm Hg (maximum feasible pressure) showed significantly less penetration of trastuzumab than in adjacent normal muscle. To establish a driving force for convection into the tumor, various maneuvers were attempted to reduce tumor pressure, including treatment with taxanes or prostaglandin E(1), elimination of tumor circulation, and removal of the tumor capsule. Tumor decapsulation decreased the pressure to zero but did not enhance the penetration of antibody. Binding to specific trastuzumab receptors on each tumor was shown to be not a significant barrier to antibody penetration. CONCLUSIONS The results only partially support our hypothesis and imply that the microenvironment of the tumor is in itself a major barrier to delivery of charged macromolecules.
Collapse
Affiliation(s)
- Michael F Flessner
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | |
Collapse
|
382
|
Binder DK, Papadopoulos MC, Haggie PM, Verkman AS. In vivo measurement of brain extracellular space diffusion by cortical surface photobleaching. J Neurosci 2005; 24:8049-56. [PMID: 15371505 PMCID: PMC6729785 DOI: 10.1523/jneurosci.2294-04.2004] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Molecular diffusion in the brain extracellular space (ECS) is an important determinant of neural function. We developed a brain surface photobleaching method to measure the diffusion of fluorescently labeled macromolecules in the ECS of the cerebral cortex. The ECS in mouse brain was labeled by exposure of the intact dura to fluorescein-dextrans (M(r) 4, 70, and 500 kDa). Fluorescein-dextran diffusion, detected by fluorescence recovery after laser-induced cortical photobleaching using confocal optics, was slowed approximately threefold in the brain ECS relative to solution. Cytotoxic brain edema (produced by water intoxication) or seizure activity (produced by convulsants) slowed diffusion by >10-fold and created dead-space microdomains in which free diffusion was prevented. The hindrance to diffusion was greater for the larger fluorescein-dextrans. Interestingly, slowed ECS diffusion preceded electroencephalographic seizure activity. In contrast to the slowed diffusion produced by brain edema and seizure activity, diffusion in the ECS was faster in mice lacking aquaporin-4 (AQP4), an astroglial water channel that facilitates fluid movement between cells and the ECS. Our results establish a minimally invasive method to quantify diffusion in the brain ECS in vivo, revealing stimulus-induced changes in molecular diffusion in the ECS with unprecedented spatial and temporal resolution. The in vivo mouse data provide evidence for: (1) dead-space ECS microdomains after brain swelling; (2) slowed molecular diffusion in the ECS as an early predictor of impending seizure activity; and (3) a novel role for AQP4 as a regulator of brain ECS.
Collapse
Affiliation(s)
- Devin K Binder
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94143-0521, USA
| | | | | | | |
Collapse
|
383
|
Tatarkova SA, Berk DA. Probing single DNA mobility with fluorescence correlation microscopy. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2005; 71:041913. [PMID: 15903707 DOI: 10.1103/physreve.71.041913] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2003] [Revised: 09/27/2004] [Indexed: 05/02/2023]
Abstract
Fluorescence correlation spectroscopy combined with microscopy (FCSM) is used to study the mobility of DNA fragments in aqueous solution and tissue models on the single molecule level. The effective hydrodynamic radius was measured for various lengths of ds-DNA chains and obeyed the theoretically inveterate [DNA length](0.5) relationship. Hindered diffusion of ds-DNA through the gel matrix of various densities is thought of as an extension of Kramer's problem for a flexible polymer chain. With increasing DNA length the average barrier crossing time rises as [DNA length](2) and this agrees with theory predictions for polymer molecules surmounting an entropic barrier.
Collapse
|
384
|
Affiliation(s)
- Bryan P Schneider
- Division of Hematology-Oncology, Indiana University, 535 Barnhill Dr, RT-473 Indianapolis, IN 46202, USA.
| | | |
Collapse
|
385
|
Miller KD, Sweeney CJ, Sledge GW. Can tumor angiogenesis be inhibited without resistance? EXS 2005:95-112. [PMID: 15617473 DOI: 10.1007/3-7643-7311-3_7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Kathy D Miller
- Division of Hematology and Oncology, Department of Medicine, Indiana University, 535 Barnhill Drive RT-473, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
386
|
Brown EB, Boucher Y, Nasser S, Jain RK. Measurement of macromolecular diffusion coefficients in human tumors. Microvasc Res 2005; 67:231-6. [PMID: 15121448 DOI: 10.1016/j.mvr.2004.02.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Indexed: 11/30/2022]
Abstract
The diffusive transport of macromolecules in tumors is an important determinant of the delivery of many anticancer therapeutics. However, measurements of diffusive transport to date have only been performed in animal models. In this work, diffusion coefficients of BSA and IgM were measured in human tumor biopsies (cooled to 4-7 degrees C to prevent degradation) using fluorescence recovery after photobleaching. To quantify the effects of excision and cooling, the diffusion coefficient of BSA and IgM was measured in human tumor xenografts in situ and after cooling and excision. The change in diffusion coefficients before and after excision of xenografts was used to calculate in vivo diffusion coefficients in human tumors from ex vivo measurements. Using this approach, we obtained the first quantitative determinations of macromolecular diffusion coefficients in human tumors and find that the diffusion coefficients of BSA and IgM in human colon were adenocarcinomas higher than those in xenografts. This difference is consistent with lower collagen content in the accessible regions of these human tumors. These measurements allow the quantitative prediction of the diffusive transport of like-sized macromolecular therapeutics in human tumors. These measurements should help in modeling the transport of novel large MW therapeutics, and hence in estimating their distribution and efficacy in tumors.
Collapse
Affiliation(s)
- Edward B Brown
- Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Boston, MA 02114, USA.
| | | | | | | |
Collapse
|
387
|
Papadopoulos MC, Binder DK, Verkman AS. Enhanced macromolecular diffusion in brain extracellular space in mouse models of vasogenic edema measured by cortical surface photobleaching. FASEB J 2004; 19:425-7. [PMID: 15596484 DOI: 10.1096/fj.04-2834fje] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Diffusion of solutes and macromolecules in brain extracellular space (ECS) is important for normal brain function and efficient drug delivery, and is thought to be impaired in edematous brain. Here we measured the diffusion of an inert macromolecular fluorescent marker (FITC-dextran, 70 kDa) in the ECS by fluorescence recovery after photobleaching after staining the exposed cerebral cortex in vivo. In a brain tumor model of vasogenic (leaky capillary) edema, FITC-dextran diffusion was reduced more than fourfold in hypercellular tumor and surrounding astrogliotic tissue; however, diffusion in brain away from the tumor was approximately 30% faster than in normal contralateral brain. The increased diffusion was abolished by dexamethasone pretreatment. Enhanced ECS diffusion was also found in uninjured brain near a region of leaky brain vessels produced by focal cortical freeze injury. In contrast, ECS diffusion was slowed more than sixfold in cytotoxic brain edema caused by anoxia. Diffusion results were related semiquantitatively to ECS volume fraction and matrix viscosity from in vitro photobleaching studies in a model system consisting of silica particles in a fluorescent water/glycerol matrix. Our data provide in vivo evidence for enhanced ECS diffusion in vasogenic brain edema, yet greatly slowed diffusion in cytotoxic edema and in and around tumors.
Collapse
Affiliation(s)
- Marios C Papadopoulos
- Department of Medicine and Physiology, University of California, San Francisco, California 94143-0521, USA
| | | | | |
Collapse
|
388
|
Eikenes L, Bruland ØS, Brekken C, Davies CDL. Collagenase increases the transcapillary pressure gradient and improves the uptake and distribution of monoclonal antibodies in human osteosarcoma xenografts. Cancer Res 2004; 64:4768-73. [PMID: 15256445 DOI: 10.1158/0008-5472.can-03-1472] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer therapy based on tumor-selective macromolecules may fail due to the elevated interstitial fluid pressure (IFP) that reduces the transvascular and interstitial convection in solid tumors. Modulation of the tumor extracellular matrix (ECM) may reduce IFP and enhance transvascular filtration and interstitial transport of macromolecules. We therefore measured the effect of the ECM-degrading enzyme collagenase on IFP and microvascular pressure (MVP) in human osteosarcoma xenografts using the wick-in-needle and micropipette methods, respectively. The tumor uptake and distribution of a systemically administered osteosarcoma-associated monoclonal antibody (TP-3) after i.v. injection of collagenase were analyzed using confocal laser scanning microscopy. Collagenase (0.1%) reduced both IFP (45%) and MVP (60%), but the kinetics of the recoveries differed, because MVP had recovered by the time IFP reached its minimum level. Thus, collagenase increased the transcapillary pressure gradient, inducing a 2-fold increase in the tumor uptake and improving the distribution of the monoclonal antibody, which was localized further into the tumor. To study the mechanism of the reduction in MVP, mean arterial blood pressure was measured and found not to be affected by the collagenase treatment. The reduction in MVP was rather due to reduced vascular resistance because microvascular-associated collagen was totally or partially disintegrated. Although collagenase may favor metastasis and thus not be clinically relevant, this study shows proof of principle that degradation of the ECM leads to a favorable change in the transvascular pressure gradient, thereby increasing antibody penetration and binding to tumor cells.
Collapse
Affiliation(s)
- Live Eikenes
- Department of Physics and Department of Circulation and Medical Imaging, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| | | | | | | |
Collapse
|
389
|
Braeckmans K, Peeters L, Sanders NN, De Smedt SC, Demeester J. Three-dimensional fluorescence recovery after photobleaching with the confocal scanning laser microscope. Biophys J 2004; 85:2240-52. [PMID: 14507689 PMCID: PMC1303450 DOI: 10.1016/s0006-3495(03)74649-9] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Confocal scanning laser microscopes (CSLMs) are equipped with the feature to photobleach user-defined regions. This makes them a handy tool to perform fluorescence recovery after photobleaching (FRAP) measurements. To allow quantification of such FRAP experiments, a three-dimensional model has been developed that describes the fluorescence recovery process for a disk-shaped geometry that is photobleached by the scanning beam of a CSLM. First the general mathematical basis is outlined describing the bleaching process for an arbitrary geometry bleached by a scanning laser beam. Next, these general expressions are applied to the bleaching by a CSLM of a disk-shaped geometry and an analytical solution is derived that describes three-dimensional fluorescence recovery in the bleached area as observed by the CSLM. The FRAP model is validated through both the Stokes-Einstein relation and the comparison of the measured diffusion coefficients with their theoretical estimates. Finally, the FRAP model is used to characterize the transport of FITC-dextrans through bulk three-dimensional biological materials: vitreous body isolated from bovine eyes, and lung sputum expectorated by cystic fibrosis patients. The decrease in the diffusion coefficient relative to its value in solution was dependent on the size of the FITC-dextrans in vitreous, whereas it was size-independent in cystic fibrosis sputum.
Collapse
Affiliation(s)
- Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | | | | | | | | |
Collapse
|
390
|
Zaharoff DA, Yuan F. Effects of pulse strength and pulse duration on in vitro DNA electromobility. Bioelectrochemistry 2004; 62:37-45. [PMID: 14990324 DOI: 10.1016/j.bioelechem.2003.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2003] [Revised: 09/25/2003] [Accepted: 10/02/2003] [Indexed: 11/17/2022]
Abstract
Interstitial transport of DNA is a rate-limiting step in electric field-mediated gene delivery in vivo. Interstitial transport of macromolecules, such as plasmid DNA, over a distance of several cell layers, is inefficient due to small diffusion coefficient and inadequate convection. Therefore, we explored electric field as a novel driving force for interstitial transport of plasmid DNA. In this study, agarose gels were used to mimic the interstitium in tissues as they had been well characterized and could be prepared reproducibly. We measured the electrophoretic movements of fluorescently labeled plasmid DNA in agarose gels with three different concentrations (1.0%, 2.0% and 3.0%) subjected to electric pulses at three different field strengths (100, 200 and 400 V/cm) and four different pulse durations (10, 50, 75, 99 ms). We observed that: (1) shorter pulses (10 ms) were not as efficient as longer pulses in facilitating plasmid transport through agarose gels; (2) plasmid electromobility reached a plateau at longer pulse durations; and (3) plasmid electromobility increased with applied electric energy, up to a threshold, in all three gels. These data suggested that both pulse strength and duration needed to be adequately high for efficient plasmid transport through extracellular matrix. We also found that electric field was better than concentration gradient of DNA as a driving force for interstitial transport of plasmid DNA.
Collapse
Affiliation(s)
- David A Zaharoff
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Durham, NC 27708, USA
| | | |
Collapse
|
391
|
Duvillard C, Ponelle T, Chapusot C, Piard F, Romanet P, Chauffert B. EDTA enhances the antitumor efficacy of intratumoral cisplatin in s.c. grafted rat colon tumors. Anticancer Drugs 2004; 15:295-9. [PMID: 15014364 DOI: 10.1097/00001813-200403000-00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We have investigated whether EDTA, a calcium chelator, could improve the accumulation of platinum in tumors and enhance the antitumor efficacy by increasing drug diffusion through the extracellular tumor matrix. Intratumoral injection of 0.3 mg/kg cisplatin combined with 10 mg/ml EDTA in 2 ml saline serum led to tumor cure in four of eight rats and produced major tumor regression in the other animals. In contrast, intratumoral injection of cisplatin alone or EDTA alone had no antitumoral effect. EDTA increased platinum accumulation both in vivo and ex vivo in the PROb tumors. EDTA alone was cytotoxic at a concentration of 10 mg/ml, but neither increased platinum accumulation nor cisplatin toxicity on cultured PROb colonic cancer cells. We conclude that EDTA could be a useful and well-tolerated adjuvant for enhancing intratumoral cisplatin chemotherapy.
Collapse
|
392
|
Alexandrakis G, Brown EB, Tong RT, McKee TD, Campbell RB, Boucher Y, Jain RK. Two-photon fluorescence correlation microscopy reveals the two-phase nature of transport in tumors. Nat Med 2004; 10:203-7. [PMID: 14716306 DOI: 10.1038/nm981] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Accepted: 12/15/2003] [Indexed: 12/19/2022]
Abstract
Transport parameters determine the access of drugs to tumors. However, technical difficulties preclude the measurement of these parameters deep inside living tissues. To this end, we adapted and further optimized two-photon fluorescence correlation microscopy (TPFCM) for in vivo measurement of transport parameters in tumors. TPFCM extends the detectable range of diffusion coefficients in tumors by one order of magnitude, and reveals both a fast and a slow component of diffusion. The ratio of these two components depends on molecular size and can be altered in vivo with hyaluronidase and collagenase. These studies indicate that TPFCM is a promising tool to dissect the barriers to drug delivery in tumors.
Collapse
Affiliation(s)
- George Alexandrakis
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
393
|
Zhou X, Pogue BW, Chen B, Hasan T. Analysis of Effective Molecular Diffusion Rates for Verteporfin in Subcutaneous Versus Orthotopic Dunning Prostate Tumors¶. Photochem Photobiol 2004; 79:323-31. [PMID: 15137508 DOI: 10.1562/mu-03-31.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Photosensitizer biodistribution change inside tissue is one of the dominant factors in photodynamic therapy efficacy. In this study, the pharmacokinetics of a benzoporphyrin derivative (BPD), delivered in verteporfin for injection formulation, have been quantified in the rat Dunning prostate tumor MAT-LyLu model, using both subcutaneous and orthotopic sites. Blood plasma sampling indicated that BPD had a bi-exponential metabolic lifetime in vivo, with the two lifetimes being 9.6 min and 8.3 h. The spatial distributions in the tumor were quantified as a function of distance from the perfused blood vessels, using fluorescence histologic images of the tumor. A fluorescent vascular marker was used to obtain locations and shapes of perfused capillaries at a wavelength of emission different from that of BPD and to allow colocalized images to be acquired of vessel and BPD locations. Using the BPD fluorescence images obtained 15 min after intravenous administration, a forward finite-element solution to the diffusion equation was used to predict the drug distribution by matching the fluorescence intensity images observed microscopically. An inverse solver was used to minimize the root mean square error between the image of simulated diffusion and the experimental image, resulting in estimation of the diffusion coefficient of BPD in the tumor models. Effective diffusion coefficients were 0.88 and 1.59 microm2/s for the subcutaneous and orthotopically grown tumors, respectively, indicating that orthotopic tumors have significantly higher vascular extravasation rates as compared with subcutaneous tumors. This analysis supports the hypothesis that leakage rates of the photosensitizer vary considerably. Thus, although varying the time between injection and optical irradiation may be used to vary the targeting between vascular and less vascular areas, the precise time of treatment will depend on the nature of the permeability of the vasculature in the tissue being treated.
Collapse
Affiliation(s)
- Xiaodong Zhou
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | | | | | | |
Collapse
|
394
|
Bamias A, Dimopoulos MA. Angiogenesis in human cancer: implications in cancer therapy. Eur J Intern Med 2003; 14:459-469. [PMID: 14962697 DOI: 10.1016/j.ejim.2003.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2003] [Accepted: 10/14/2003] [Indexed: 12/13/2022]
Abstract
Angiogenesis represents an essential step in tumor proliferation, expansion, and metastasis. Tumor cells may express both proangiogenic and/or antiangiogenic factors. Under normal circumstances, angiogenesis is controlled through the equilibrium of these factors. This balance is disrupted in malignancy, resulting in promotion of angiogenesis. Among angiogenic molecules, VEGF appears to have a central role in the angiogenic process: it is the target of many proangiogenic factors, but it also regulates molecules that are implicated in endothelial proliferation. It has been suggested that VEGF may be a proximate angiogenic factor through which others act. The degree of angiogenesis and the expression of angiogenic factors have been associated with prognosis in several human neoplasms. In addition, angiogenesis offers a theoretically selective target for anticancer therapy, since it is only required for wound healing, endometrial proliferation, and pregnancy in healthy individuals. Antiangiogenic cancer treatment is still largely experimental and its clinical potential is currently being studied in clinical trials. Thalidomide, a drug with antiangiogenic properties, has shown significant efficacy in patients with relapsed or refractory multiple myeloma. In addition, an anti-VEGF monoclonal antibody prolonged survival in patients with advanced colorectal and renal cell carcinoma. Although these results are encouraging, selection of patients is essential in order to target populations most likely to benefit from antiangiogenic therapy.
Collapse
Affiliation(s)
- Aristotle Bamias
- Department of Clinical Therapeutics, School of Medicine, University of Athens, Medical School, 31 Komninon Street, Haidari, 124 62 Athens, Greece
| | | |
Collapse
|
395
|
Voges J, Reszka R, Gossmann A, Dittmar C, Richter R, Garlip G, Kracht L, Coenen HH, Sturm V, Wienhard K, Heiss WD, Jacobs AH. Imaging-guided convection-enhanced delivery and gene therapy of glioblastoma. Ann Neurol 2003; 54:479-87. [PMID: 14520660 DOI: 10.1002/ana.10688] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In a prospective phase I/II clinical study, we treated eight patients suffering from recurrent glioblastoma multiform with stereotactically guided intratumoral convection-enhanced delivery of an HSV-1-tk gene-bearing liposomal vector and systemic ganciclovir. Noninvasive identification of target tissue together with assessment of vector-distribution volume and the effects of gene therapy were achieved using magnetic resonance imaging and positron emission tomography. The treatment was tolerated well without major side effects. In two of eight patients, we observed a greater than 50% reduction of tumor volume and in six of eight patients focal treatment effects. Intracerebral infusion of contrast medium before vector application displayed substantial inhomogeneity of tissue staining indicating the need of test infusions to monitor the mechanical distribution of vectors. Visualization of therapeutic effects on tumor metabolism and documentation of gene expression using positron emission tomography indicated that molecular imaging technology appears to be essential for the further development of biological treatment strategies.
Collapse
Affiliation(s)
- Juergen Voges
- Department of Stereotaxy and Functional Neurosurgery, University of Köln, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
396
|
Jain RK. Angiogenesis and lymphangiogenesis in tumors: insights from intravital microscopy. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:239-48. [PMID: 12858546 DOI: 10.1101/sqb.2002.67.239] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- R K Jain
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
397
|
Wildiers H, Guetens G, De Boeck G, Verbeken E, Landuyt B, Landuyt W, de Bruijn EA, van Oosterom AT. Effect of antivascular endothelial growth factor treatment on the intratumoral uptake of CPT-11. Br J Cancer 2003; 88:1979-86. [PMID: 12799646 PMCID: PMC2741115 DOI: 10.1038/sj.bjc.6601005] [Citation(s) in RCA: 266] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Promising preclinical activity with agents blocking the function of vascular endothelial growth factor (VEGF) has been observed in various cancer types, especially with combination therapy. However, these drugs decrease microvessel density, and it is not known whether this reduced vessel density (VD) results in decreased delivery of concomitantly administered classical anticancer drugs. We designed an in vivo study to investigate the relation between VEGF-blocking therapy, tumoral blood vessels, and intratumoral uptake of anticancer drugs. Nude NMRI mice bearing colon adenocarcinoma (HT29) were treated with the anti-VEGFmAb A4.6.1 or placebo. After 1 week, CPT-11 was administered 1 h prior to killing the animals. In A4.6.1 treated tumours, there was a significant decrease in VD, more pronounced with potentially functional large vessels than endothelial cords. Interestingly, a trend to increased intratumoral CPT-11 concentration was observed (P=0.09). In parallel, we measured an increase in tumour perfusion, as estimated by high-performance liquid chromatography determination of intratumoural Hoechst 33342 concentration. In the growth delay study, CPT-11 was at least equally effective with or without pretreatment with A4.6.1. These data suggest that tumour vascular function and tumour uptake of anticancer drugs improve with VEGF-blocking therapy, and indicate the relevance for further investigations.
Collapse
Affiliation(s)
- H Wildiers
- Laboratory of Experimental Oncology (LEO), University Hospital Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
398
|
Brown E, McKee T, diTomaso E, Pluen A, Seed B, Boucher Y, Jain RK. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat Med 2003; 9:796-800. [PMID: 12754503 DOI: 10.1038/nm879] [Citation(s) in RCA: 568] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2002] [Accepted: 04/25/2003] [Indexed: 12/13/2022]
Abstract
The content and structure of collagen is essential in governing the delivery of therapeutic molecules in tumors. Thus, simple histological staining of tumor tissue biopsies for collagen could be used to assess the accessibility of molecular therapeutics in tumors. Here we show that it is possible to optically image fibrillar collagen in tumors growing in mice using second-harmonic generation (SHG). Using this noninvasive technique, we estimated relative diffusive hindrance, quantified the dynamics of collagen modification after pharmacologic intervention and provided mechanistic insight into improved diffusive transport induced by the hormone relaxin. This technology could offer basic scientists and clinicians an enhanced ability to estimate the relative penetrabilities of molecular therapeutics.
Collapse
Affiliation(s)
- Edward Brown
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
399
|
Shan S, Sorg B, Dewhirst MW. A novel rodent mammary window of orthotopic breast cancer for intravital microscopy. Microvasc Res 2003; 65:109-17. [PMID: 12686168 DOI: 10.1016/s0026-2862(02)00017-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Orthotopic and ectopic organ environments differentially influence tumor growth, metastasis, and sensitivity to therapy. In this study we present a novel rodent mammary window of orthotopic breast cancer, which is amenable to study of microvascular function and angiogenesis in this orthotopic site. The skin around the nipple of selected mammary glands of female Fischer 344 rats was removed and the nipple was cut at its base. R3230Ac tumor fragments or cells in Gelfoam were aseptically implanted into the nipple sinus. An acrylic disk was placed on top of the implant and was sutured in place. Histology showed that tumors were well established within 5 days. Similar techniques were also applied to BALB/c mice transplanted with 4T1 murine mammary carcinoma cells. With GFP-expressing tumor cells and serial observations, we demonstrated unique patterns of tumor cell proliferation and vascularization in both tumor models. The images obtained were comparable to those from the dorsal skinfold window chambers. This model will allow for study of tumor microcirculatory function, angiogenesis, tumor cell-host interactions, and evaluation of effects of various treatments.
Collapse
Affiliation(s)
- Siqing Shan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
400
|
Miller KD, Sweeney CJ, Sledge GW. The Snark is a Boojum: the continuing problem of drug resistance in the antiangiogenic era. Ann Oncol 2003; 14:20-8. [PMID: 12488288 DOI: 10.1093/annonc/mdg033] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
If your Snark be a Snark, that is right: Fetch it home by all means-you may serve it with greens, And it's handy for striking a light. "But oh, beamish nephew, beware of the day, If your Snark be a Boojum! For then You will softly and suddenly vanish away, And never be met with again!" Lewis Carroll The Hunting of the Snark
Collapse
Affiliation(s)
- K D Miller
- Division of Hematology and Oncology, Indiana University, Indianapolis, USA.
| | | | | |
Collapse
|