Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 6: e1815, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26158521

ABSTRACT

Approved proteasome inhibitors have advanced the treatment of multiple myeloma but are associated with serious toxicities, poor pharmacokinetics, and most with the inconvenience of intravenous administration. We therefore sought to identify novel orally bioavailable proteasome inhibitors with a continuous daily dosing schedule and improved therapeutic window using a unique drug discovery platform. We employed a fluorine-based medicinal chemistry technology to synthesize 14 novel analogs of epoxyketone-based proteasome inhibitors and screened them for their stability, ability to inhibit the chymotrypsin-like proteasome, and antimyeloma activity in vitro. The tolerability, pharmacokinetics, pharmacodynamic activity, and antimyeloma efficacy of our lead candidate were examined in NOD/SCID mice. We identified a tripeptide epoxyketone, FV-162, as a metabolically stable, potent proteasome inhibitor cytotoxic to human myeloma cell lines and primary myeloma cells. FV-162 had limited toxicity and was well tolerated on a continuous daily dosing schedule. Compared with the benchmark oral irreversible proteasome inhibitor, ONX-0192, FV-162 had a lower peak plasma concentration and longer half-life, resulting in a larger area under the curve (AUC). Oral FV-162 treatment induced rapid, irreversible inhibition of chymotrypsin-like proteasome activity in murine red blood cells and inhibited tumor growth in a myeloma xenograft model. Our data suggest that oral FV-162 with continuous daily dosing schedule displays a favorable safety, efficacy, and pharmacokinetic profile in vivo, identifying it as a promising lead for clinical evaluation in myeloma therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Fluorine/administration & dosage , Multiple Myeloma/drug therapy , Oligopeptides/administration & dosage , Proteasome Inhibitors/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Fluorine/pharmacokinetics , Humans , Mice , Multiple Myeloma/pathology , Proteasome Inhibitors/pharmacokinetics , Xenograft Model Antitumor Assays
2.
Conf Proc IEEE Eng Med Biol Soc ; 2004: 1321-4, 2004.
Article in English | MEDLINE | ID: mdl-17271935

ABSTRACT

Current density impedance imaging (CDII) is a new impedance imaging technique that utilizes current density vector measurements made using magnetic resonance imager (MRI). CDII provides a simple mathematical expression for the gradient of the logarithm of conductivity, nablaln(sigma), at each point in a region where two current density vector has been measured. From the images of the gradient of the logarithm of conductivity, ln(sigma) can be reconstructed through integration and of sigma by a priori knowledge of the conductivity at a single point in the object. The CDII technique was tested on a conductivity phantom made from tissue mimicking gel. The results showed accurate reconstruction of the gel conductivity from two current density measurements. This study, for the first time, has demonstrated a local reconstruction technique to calculate sample conductivity inside the phantom noninvasively.

3.
Article in English | MEDLINE | ID: mdl-11370348

ABSTRACT

Large-scale simulation of ultrasonic pulse propagation in inhomogeneous tissue is important for the study of ultrasound-tissue interaction as well as for development of new imaging methods. Typical scales of interest span hundreds of wavelengths; most current two-dimensional methods, such as finite-difference and finite-element methods, are unable to compute propagation on this scale with the efficiency needed for imaging studies. Furthermore, for most available methods of simulating ultrasonic propagation, large-scale, three-dimensional computations of ultrasonic scattering are infeasible. Some of these difficulties have been overcome by previous pseudospectral and k-space methods, which allow substantial portions of the necessary computations to be executed using fast Fourier transforms. This paper presents a simplified derivation of the k-space method for a medium of variable sound speed and density; the derivation clearly shows the relationship of this k-space method to both past k-space methods and pseudospectral methods. In the present method, the spatial differential equations are solved by a simple Fourier transform method, and temporal iteration is performed using a k-t space propagator. The temporal iteration procedure is shown to be exact for homogeneous media, unconditionally stable for "slow" (c(x) < or = c0) media, and highly accurate for general weakly scattering media. The applicability of the k-space method to large-scale soft tissue modeling is shown by simulating two-dimensional propagation of an incident plane wave through several tissue-mimicking cylinders as well as a model chest wall cross section. A three-dimensional implementation of the k-space method is also employed for the example problem of propagation through a tissue-mimicking sphere. Numerical results indicate that the k-space method is accurate for large-scale soft tissue computations with much greater efficiency than that of an analogous leapfrog pseudospectral method or a 2-4 finite difference time-domain method. However, numerical results also indicate that the k-space method is less accurate than the finite-difference method for a high contrast scatterer with bone-like properties, although qualitative results can still be obtained by the k-space method with high efficiency. Possible extensions to the method, including representation of absorption effects, absorbing boundary conditions, elastic-wave propagation, and acoustic nonlinearity, are discussed.


Subject(s)
Models, Biological , Ultrasonography/statistics & numerical data , Adipose Tissue/diagnostic imaging , Algorithms , Biomedical Engineering , Humans , Scattering, Radiation
4.
In Vitro Cell Dev Biol Anim ; 36(6): 367-73, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10949995

ABSTRACT

Our hypothesis is that rotation increases apoptosis in standard tissue culture medium at shear stresses of greater than approximately 0.3 dyn/cm2. Human MIP-101 poorly differentiated colorectal carcinoma cells were cultured for 6 d in complete medium in monolayers, on Teflon-coated nonadherent surfaces (static three-dimensional [3D]) or in rotating 3D cultures either in microgravity in low-earth orbit (3D microg) or in unit gravity on the ground (3D 1g). Apoptosis (determined morphologically), proliferation (by MIB1 staining), and the expression of epidermal growth-factor receptor (EGF-R), TGF-alpha, or TGF-beta were assessed by immunohistochemistry, while the expression of the differentiation marker carcinoembryonic antigen (CEA) was assessed on Western blots. Over the course of 6 d, static 3D cultures displayed the highest rates of proliferation and lowest apoptosis. This was associated with high EGF-R, TGF-alpha, and TGF-beta expression which was greater than that of a monolayer culture. Both rotated 3D lg and 3D microg cultures displayed lower expression of EGF-R, TGF-alpha, or TGF-beta and proliferation than that of monolayer or static 3D cultures. However, rotated 3D microg displayed significantly less apoptosis and greater CEA expression than rotated 3D 1g cultures. When rotated cultures of MIP-101 cells were grown uncler static conditions for another 3 d, proliferation increased and apoptosis decreased. Thus, rotation appears to increase apoptosis and decrease proliferation, whereas static 3D cultures in either unit or microgravity have less apoptosis, and reduced rotation in microgravity increases CEA expression.


Subject(s)
Apoptosis , Cell Culture Techniques , Cell Differentiation , Colorectal Neoplasms/pathology , Tumor Cells, Cultured/cytology , Weightlessness , Bioreactors , Carcinoembryonic Antigen/analysis , Cell Adhesion , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Division , Cell Size , Colorectal Neoplasms/chemistry , Culture Media , ErbB Receptors/analysis , Humans , Rotation , Transforming Growth Factor alpha/analysis , Transforming Growth Factor beta/analysis , Tumor Cells, Cultured/chemistry
5.
Cancer Res ; 58(7): 1524-31, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537259

ABSTRACT

Human colorectal carcinoma (CRC) cell survival for the first 24 h after implantation in the hepatic sinusoid determines its potential to colonize the liver. Nearly 10-fold more highly metastatic CX-1 cells survive within the livers of nude mice 24 h after intrasplenic injection than weakly metastatic clone A cells. Because CRCs contact sinusoidal endothelial cells (SECs) during implantation, we sought to determine whether SECs were more toxic to clone A than to CX-1 cells. When 2 x 10(4) vital dye-labeled CRC cells were added to murine SEC monolayers, more than 30% of clone A cells lost calcein AM fluorescence compared to fewer than 5% of CX-1 cells after 24 h of coculture with SECs. Kupffer cells did not mediate this effect, because neither enriched Kupffer cells nor SECs treated with a Kupffer cell inhibitor altered the SEC-mediated toxic effect to clone A cells. Pretreatment with a nitric oxide synthase inhibitor, N(G)-monomethyl-L-arginine, superoxide dismutase, or dexamethasone, blocked SEC-mediated toxicity to clone A cells, whereas calcium chelation and catalase did not. In addition, clone A cells were more sensitive to a superoxide donor, 3-morpholinosydnonimine N-ethylcarbamide, than were CX-1 cells, and neither cell line was sensitive to sodium nitroprusside, a nitric oxide donor. Thus, unstimulated murine SECs produce reactive oxygen species that are selectively toxic to weakly metastatic clone A cells. This may be a mechanism by which host liver cells eliminate weakly metastatic neoplastic cells.


Subject(s)
Carcinoma/pathology , Cell Communication/physiology , Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Liver/cytology , Nitric Oxide/physiology , Superoxides/metabolism , Animals , Carcinoma/metabolism , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Colorectal Neoplasms/metabolism , Endothelium/cytology , Endothelium/metabolism , Enzyme Inhibitors/pharmacology , Humans , Liver/metabolism , Male , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Superoxides/antagonists & inhibitors , Tumor Cells, Cultured
6.
Cancer Res ; 57(19): 4432-6, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9331108

ABSTRACT

Carcinoembryonic antigen (CEA) may promote experimental metastasis through production of cytokines. The effect of systemic CEA on the production of proinflammatory cytokines was investigated in mice and compared to levels induced by lipopolysaccharide (LPS). Serum concentrations of interleukin (IL)-6 peaked 1 h after an i.v. CEA injection of 40 microg/mouse to 37-54% of the maximal level induced by a 1 microg/mouse injection of LPS in both normal and immunoincompetent mice. The CEA induction of IL-6 was a specific response, because the peptide PELPK (the pentapeptide on CEA that is the ligand for the CEA receptor on Kupffer cells) conjugated to albumin induced 30% of the maximal CEA response for IL-6, whereas the specificity control PELGK-conjugated albumin did not. IL-1alpha and tumor necrosis factor (TNF)-alpha levels after i.v. injection of CEA were only 3-5% of those induced by LPS. The IL-6 responses of mice pretreated with 100 microg/kg genistein were decreased by more than 40%. However, genistein inhibited the TNF-alpha response to LPS by 46% but increased the CEA-induced response by 300%. When murine Kupffer cells were stimulated with LPS or CEA in vitro, LPS increased tyrosine phosphorylation of a Mr 30,000 protein, whereas CEA decreased phosphorylation of a Mr 60,000 protein and did not increase phosphorylation of the Mr 30,000 protein. Thus, i.v. CEA stimulates production of IL-6 and TNF-alpha after binding to Kupffer cells through signal transduction pathways that appear to be different from those stimulated by LPS.


Subject(s)
Carcinoembryonic Antigen/pharmacology , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Animals , Carcinoembryonic Antigen/chemistry , Cytokines/blood , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Interleukin-1/blood , Interleukin-6/blood , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/analysis
7.
J Acoust Soc Am ; 102(2 Pt 1): 715-25, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9265750

ABSTRACT

An inverse scattering method that uses eigenfunctions of the scattering operator is presented. This approach provides a unified framework that encompasses eigenfunction methods of focusing and quantitative image reconstruction in arbitrary media. Scattered acoustic fields are described using a compact, normal operator. The eigenfunctions of this operator are shown to correspond to the far-field patterns of source distributions that are directly proportional to the position-dependent contrast of a scattering object. Conversely, the eigenfunctions of the scattering operator specify incident-wave patterns that focus on these effective source distributions. These focusing properties are employed in a new inverse scattering method that represents unknown scattering media using products of numerically calculated fields of eigenfunctions. A regularized solution to the nonlinear inverse scattering problem is shown to result from combinations of these products, so that the products comprise a natural basis for efficient and accurate reconstructions of unknown inhomogeneities. The corresponding linearized problem is solved analytically, resulting in a simple formula for the low-pass-filtered scattering potential. The linear formula is analytically equivalent to known filtered-backpropagation formulas for Born inversion, and, at least in the case of small scattering objects, has advantages of computational simplicity and efficiency. A similarly efficient and simple formula is derived for the nonlinear problem in which the total acoustic pressure can be determined based on an estimate of the medium. Computational results illustrate focusing of eigenfunctions on discrete and distributed scattering media, quantitative imaging of inhomogeneous media using products of retransmitted eigenfunctions, inverse scattering in an inhomogeneous background medium, and reconstructions for data corrupted by noise.


Subject(s)
Sound , Humans , Models, Theoretical
8.
In Vitro Cell Dev Biol Anim ; 33(5): 352-7, 1997 May.
Article in English | MEDLINE | ID: mdl-9196893

ABSTRACT

MIP-101 is a poorly differentiated human colon carcinoma cell line established from ascites that produces minimal amounts of carcinoembryonic antigen (CEA), a 180 kDa glycoprotein tumor marker, and nonspecific cross-reacting antigen (NCA), a related protein that has 50 and 90 kDa isoforms, in monolayer culture. However, MIP-101 produces CEA when implanted into the peritoneum of nude mice but not when implanted into subcutaneous tissue. We tested whether three-dimensional (3D) growth was a sufficient stimulus to produce CEA and NCA 50/90 in MIP-101 cells, because cells grow in 3D in vivo rather than in two-dimensions (2D) as occurs in monolayer cultures. To do this, MIP-101 cells were cultured on microcarrier beads in 3D cultures, either in static cultures as nonadherent aggregates or under dynamic conditions in a NASA-designed low shear stress bioreactor. MIP-101 cells proliferated well under all three conditions and increased CEA and NCA production three- to four-fold when grown in 3D cultures compared to MIP-101 cells growing logarithmically in monolayers. These results suggest that 3D growth in vitro simulates tumor function in vivo and that 3D growth by itself may enhance production of molecules that are associated with the metastatic process.


Subject(s)
Antigens, Neoplasm , Carcinoembryonic Antigen/biosynthesis , Cell Adhesion Molecules , Membrane Glycoproteins/biosynthesis , Animals , Bioreactors , Cell Division , Cytoplasm/metabolism , Humans , Mice , Mice, Nude , Tumor Cells, Cultured
9.
Clin Exp Metastasis ; 14(2): 153-64, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8605729

ABSTRACT

In vivo fluorescence videomicroscopy (IVFM) was used to analyse the behavior of weakly and highly metastatic human colorectal carcinoma (CRC) cells during implantation in the liver. A highly metastatic human CRC cell line, CX-1, and a weakly metastatic line, Clone A, were double-labeled with rhodamine B isothiocyanate-dextran (Rd-Dx) to locate cells and with calcein AM to assess cell metabolic activity in an experimental metastasis model. Double-labeled CRC cells (2.0 x 10(6)) were injected into the spleens of groups of nude mice and the livers observed by IVFM over the next 72 h. CRC cells were implanted within 30 s after injection into either portal venules or the proximal third of hepatic sinusoids. Approximately 0.5% of CRC cells traversed the liver through portal-central venous shunts and implanted in the lung. The number of CX-1 cells in the liver was similar to that of Clone A cells during the first 12 h. However, more CX-1 cells than Clone A cells remained in the liver at 4 h and were in groups of 8-12 cells whereas only a few, single Clone A cells were detected in the liver at 72 h. Not all Clone A cells are committed to die within 4 h of implantation because cells harvested 4 h after hepatic implantation proliferated normally in vitro when removed from the hepatic microenvironment. Since the stress of mechanical deformation during implantation may cause differences in cell survival, CX-1 and Clone A cells were passed through filters with 8 microM pores in vitro at 10-15 cm of water pressure to recreate the trauma of hepatic implantation. Approximately 50% of both CX-1 and Clone A cells were lysed. Furthermore, both CRC lines remained metabolically active when co-cultivated with liver cells for at least 24 h in vitro. Thus, the difference in metastatic potential between the two CRC lines may reside in their response to the combination of mechanical implantation and subsequent growth in the liver parenchyma.


Subject(s)
Carcinoma/pathology , Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Animals , Cell Adhesion , Cell Survival , Clone Cells , Fluorescent Dyes , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Stress, Mechanical , Time Factors , Tumor Cells, Cultured , Video Recording
10.
Surg Oncol ; 2(4): 255-64, 1993 Aug.
Article in English | MEDLINE | ID: mdl-7504563

ABSTRACT

CD44 is a cellular adhesion molecule expressed in many different types of cells that may be a receptor for hyaluronic acid, laminin, collagen or fibronectin. In this study, we determined whether CD44 participated in the adhesion of three human colorectal carcinoma cell lines (KM-12c, CCL 188 and MIP-101) to laminin, collagen and hyaluronic acid. All lines were positive for the epithelial form of CD44 (CD44E) with a molecular weight of approximately 160 kD. All of them bound significantly to laminin and type IV collagen but not to hyaluronic acid in a solid phase adhesion assay. Three monoclonal antibodies to CD44 (Hermes 1, Hermes 3 and J173) significantly blocked the binding of colorectal carcinoma lines to laminin and collagen whereas another antibody to CD44 (50B4) bound to cells but did not inhibit adhesion. Only Hermes 1 completely abolished the binding to hyaluronic acid by a human B lymphoblastoid cell line, JY, that expressed the 90 kD haematopoietic form of CD44. Soluble hyaluronate inhibited the adhesion of JY cells to solid phase hyaluronate but did not inhibit adhesion to laminin and collagen by the colorectal carcinoma lines. Thus, (a) CD44E participates in the adhesion of colorectal carcinoma cells to laminin and type IV collagen and (b) the binding site for laminin and collagen on CD44E is different from the site for hyaluronic acid.


Subject(s)
Carcinoma/metabolism , Collagen/metabolism , Colorectal Neoplasms/metabolism , Laminin/metabolism , Receptors, Lymphocyte Homing/physiology , Antibodies, Monoclonal/immunology , Carcinoma/immunology , Cell Adhesion , Colorectal Neoplasms/immunology , Fluorescent Antibody Technique , Humans , Hyaluronan Receptors , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Immunoblotting , Metalloendopeptidases/pharmacology , Receptors, Lymphocyte Homing/analysis , Receptors, Lymphocyte Homing/immunology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL