Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Sci Rep ; 10(1): 9655, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32541845

ABSTRACT

Intervertebral disc (IVD) degeneration with chronic low back pain is associated with neo-vascularisation into the deeper IVD regions. During this process, endothelial cells (ECs), which are primarily responsible for angiogenesis, interact with the adjacent annulus fibrosus (AF) cells, which are the first line of defence against the invasion of vascular structures into deeper IVD regions. However, the accumulation of inflammatory and catabolic enzymes that results from this interaction promotes matrix degradation and an inflammatory response. Thus, regulating the production of these mediators and catabolic enzymes could ameliorate IVD degeneration. Photobiomodulation (PBM) therapy is a non-invasive stimulation known to have biologically beneficial effects on wound healing, tissue repair, and inflammation. Here, we examined the effects of PBM, administered at various wavelengths (645, 525, and 465 nm) and doses (16, 32, and 64 J/cm2), on EC-stimulated human AF cells. Our results show that PBM selectively inhibited the EC-mediated production of inflammatory mediators, catabolic enzymes, and neurotrophins by human AF cells in a dose- and wavelength-dependent manner. These results suggest that PBM could be a superior and advanced treatment strategy for IVD degeneration.


Subject(s)
Annulus Fibrosus/cytology , Culture Media, Conditioned/chemistry , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Intervertebral Disc Degeneration/metabolism , Low-Level Light Therapy/methods , Neovascularization, Pathologic/metabolism , Adult , Annulus Fibrosus/metabolism , Annulus Fibrosus/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Endothelial Cells/chemistry , Extracellular Matrix/genetics , Female , Gene Expression Regulation/radiation effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/radiotherapy , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Middle Aged , Models, Biological , Neovascularization, Pathologic/radiotherapy
2.
Eur J Nucl Med Mol Imaging ; 45(1): 123-131, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28821931

ABSTRACT

PURPOSE: Increased angiogenesis is a marker of aggressiveness in many cancers. Targeted radionuclide therapy of these cancers with angiogenesis-targeting agents may curtail this increased blood vessel formation and slow the growth of tumors, both primary and metastatic. CD105, or endoglin, has a primary role in angiogenesis in a number of cancers, making this a widely applicable target for targeted radioimmunotherapy. METHODS: The anti-CD105 antibody, TRC105 (TRACON Pharmaceuticals), was conjugated with DTPA for radiolabeling with 177Lu (t 1/2 6.65 days). Balb/c mice were implanted with 4T1 mammary carcinoma cells, and five study groups were used: 177Lu only, TRC105 only, 177Lu-DTPA-IgG (a nonspecific antibody), 177Lu-DTPA-TRC105 low-dose, and 177Lu-DTPA-TRC105 high-dose. Toxicity of the agent was monitored by body weight measurements and analysis of blood markers. Biodistribution studies of 177Lu-DTPA-TRC105 were also performed at 1 and 7 days after injection. Ex vivo histology studies of various tissues were conducted at 1, 7, and 30 days after injection of high-dose 177Lu-DTPA-TRC105. RESULTS: Biodistribution studies indicated steady uptake of 177Lu-DTPA-TRC105 in 4T1 tumors between 1 and 7 days after injection (14.3 ± 2.3%ID/g and 11.6 ± 6.1%ID/g, respectively; n = 3) and gradual clearance from other organs. Significant inhibition of tumor growth was observed in the high-dose group, with a corresponding significant increase in survival (p < 0.001, all groups). In most study groups (all except the nonspecific IgG group), the body weights of the mice did not decrease by more than 10%, indicating the safety of the injected agents. Serum alanine transaminase levels remained nearly constant indicating no damage to the liver (a primary clearance organ of the agent), and this was confirmed by ex vivo histological analyses. CONCLUSION: 177Lu-DTPA-TRC105, when administered at a sufficient dose, is able to curtail tumor growth and provide a significant survival benefit without off-target toxicity. Thus, this targeted agent could be used in combination with other treatment options to slow tumor growth allowing the other agents to be more effective.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lutetium/chemistry , Neoplasms, Experimental/radiotherapy , Neovascularization, Pathologic/radiotherapy , Radioimmunotherapy/methods , Radioisotopes/chemistry , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Endoglin/immunology , Female , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Pentetic Acid/chemistry , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
3.
Sci Rep ; 7: 40243, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28074839

ABSTRACT

Antivascular therapy represents a proven strategy to treat angiogenesis. By applying synchronized ultrasound bursts and nanosecond laser irradiation, we developed a novel, selective, non-invasive, localized antivascular method, termed photo-mediated ultrasound therapy (PUT). PUT takes advantage of the high native optical contrast among biological tissues and can treat microvessels without causing collateral damage to the surrounding tissue. In a chicken yolk sac membrane model, under the same ultrasound parameters (1 MHz at 0.45 MPa and 10 Hz with 10% duty cycle), PUT with 4 mJ/cm2 and 6 mJ/cm2 laser fluence induced 51% (p = 0.001) and 37% (p = 0.018) vessel diameter reductions respectively. With 8 mJ/cm2 laser fluence, PUT would yield vessel disruption (90%, p < 0.01). Selectivity of PUT was demonstrated by utilizing laser wavelengths at 578 nm or 650 nm, where PUT selectively shrank veins or occluded arteries. In a rabbit ear model, PUT induced a 68.5% reduction in blood perfusion after 7 days (p < 0.001) without damaging the surrounding cells. In vitro experiments in human blood suggested that cavitation may play a role in PUT. In conclusion, PUT holds significant promise as a novel non-invasive antivascular method with the capability to precisely target blood vessels.


Subject(s)
Low-Level Light Therapy , Neovascularization, Pathologic/radiotherapy , Ultrasonic Therapy , Animals , Blood/radiation effects , Chickens , Ear/blood supply , Ear/radiation effects , Humans , Rabbits , Yolk Sac/blood supply , Yolk Sac/radiation effects
4.
Technol Health Care ; 24(5): 607-26, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27129030

ABSTRACT

Photothermal treatment of tumors of the retina and choroid such as retinoblastomas, malignant melanomas, benign tumors as well as of vascular malformations can be performed by using laser radiation. A number of basic physical laws have to be taken into account in this procedure. Of particular importance thereby are: Arrhenius' law to approximate the kinetics of protein denaturation and photocoagulation, furthermore the electromagnetic radiation field, the distribution of both radiant and thermal energy induced in tumors and vascular structures, the influence of the wavelength and laser pulse duration (exposure time), as well as of the optical properties of the tissue. Strict confinement of the extent of the photothermal damage is critical since such pathological entities are frequently located close to the macula or optic nerve head.The conditions for tumor destruction are best fulfilled when using radiation in the near-infrared range of the electromagnetic spectrum such as that emitted from the diode (810 nm) and the Nd: YAG (1064 nm) laser, because of the good optical penetration properties of these radiations in tissue. Short wavelength sources of radiation, such as the argon ion (488, 514 nm) or the freqeuency-doubled Nd: YAG (532 nm) laser are less well suited for the irradiation of large vascular structures due to their poor penetration depths. However, for vascular formations with a small thickness (1 mm or less), short wavelength sources appear to be the most appropriate choice. Optical coupling of radiant energy to the eye by means of indirect ophthalmoscopic systems or positive contact lenses is furthermore of importance. Strong positive lenses may lead to severe constrictions of the laser beam within the anterior segment, that leads to high irradiance increasing the probability for structures to be damaged; with negative contact lenses, such as the -64 D Goldmann type lens, this danger is largely absent.


Subject(s)
Choroid Neoplasms/radiotherapy , Low-Level Light Therapy/methods , Phototherapy/methods , Retinal Neoplasms/radiotherapy , Hemangioma/radiotherapy , Lasers, Solid-State , Neovascularization, Pathologic/radiotherapy , Protein Denaturation/radiation effects
5.
J Therm Biol ; 44: 55-62, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25086974

ABSTRACT

Laser-induced hyperthermia treatment of tumor in a 2-D axisymmetric tissue embedded with moderate size (100-150µm) blood vessels is studied. Laser absorption is enhanced by embedding gold-silica nanoshells in the tumor. Heat transfer in the tissue is modeled using Weinbaum-Jiji bioheat transfer equation. With laser irradiation, the volumetric radiation is accounted in the governing bioheat equation. Radiative information needed in the bioheat equation is calculated using the discrete ordinate method, and the coupled bioheat-radiation equation is solved using the finite volume method. Effects of power density, laser exposure time, beam radius, diameter of blood vessel and volume fractions of nanoshells on temperature spread in the tissue are analyzed.


Subject(s)
Low-Level Light Therapy , Models, Biological , Neovascularization, Pathologic/radiotherapy , Animals , Gold/chemistry , Humans , Nanoshells/chemistry
6.
PLoS One ; 9(1): e86421, 2014.
Article in English | MEDLINE | ID: mdl-24475118

ABSTRACT

BACKGROUND AND AIM: Recurrence and metastasis are associated with poor prognosis in hepatocellular carcinoma even in the patients who have undergone radical resection. Therefore, effective treatment is urgently needed for improvement of patients' survival. Previously, we reported that nanosecond pulse electric fields (nsPEFs) can ablate melanoma by induction of apoptosis and inhibition of angiogenesis. This study aims to investigate the in vivo ablation strategy by comparing the dose effect of nanosecond electric fields in vitro and in vivo on hepatocellular carcinoma. MATERIALS AND METHODS: Four hepatocellular carcinoma cell lines HepG2, SMMC7721, Hep1-6, and HCCLM3 were pulsed to test the anti-proliferation and anti-migration ability of 100 ns nsPEFs in vitro. The animal model of human subdermal xenograft HCCLM3 cells into BALB/c nude mouse was used to test the anti-tumor growth and macrophage infiltration in vivo. RESULTS: In vitro assays showed anti-tumor effect of nsPEFs is dose-dependant. But the in vivo study showed the strategy of low dose and multiple treatments is superior to high dose single treatment. The macrophages infiltration significantly increased in the tumors which were treated by multiple low dose nsPEFs. CONCLUSION: The low dose multiple nsPEFs application is more efficient than high dose single treatment in inhibiting the tumor volume in vivo, which is quite different from the dose-effect relationship in vitro. Beside the electric field strength, the macrophage involvement must be considered to account for effect variability and toxicology in vivo.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/radiotherapy , Electric Stimulation Therapy/methods , Liver Neoplasms/immunology , Liver Neoplasms/radiotherapy , Macrophage Activation/immunology , Analysis of Variance , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/radiotherapy
7.
Radiology ; 257(2): 342-52, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20829544

ABSTRACT

PURPOSE: To evaluate the sequential injection of a low-molecular-weight (gadoterate meglumine [Gd-DOTA], 0.5 kDa) and a macromolecular (P846, 3.5 kDa) contrast media in monitoring the effect of antitumor therapies (antiangiogenic therapy and/or microbeam radiation therapy [MRT]) on healthy brain tissue and implanted tumors. MATERIALS AND METHODS: Animal use was compliant with official French guidelines and was assessed by the local Internal Evaluation Committee for Animal Welfare and Rights. Eighty male rats bearing 9L gliosarcoma were randomized into four groups: untreated, antiangiogenic (sorafenib) therapy, MRT, and both treatments. Magnetic resonance (MR) imaging was performed 1 day before and 1, 5, and 8 days after the start of the treatment. At all time points, vascular integrity to a macromolecular contrast medium (P846) and, 11 minutes 30 seconds later, to low-molecular-weight contrast medium (Gd-DOTA) was evaluated by using a dynamic contrast material-enhanced MR imaging approach. To quantify vessel wall integrity, areas under the signal intensity curves were computed for each contrast medium. Unpaired t tests and one-way analysis of variance were used for statistical analyses. RESULTS: Tumor vessels receiving antiangiogenic therapy became less permeable to the macromolecular contrast medium, but their permeability to the low-molecular-weight contrast medium remained unchanged. Healthy double-irradiated vessels became permeable to the low-molecular-weight contrast medium but not to the macromolecular contrast medium. CONCLUSION: Antiangiogenic therapy and MRT generate different effects on the extravasation of contrast medium in tumoral and healthy tissues. This study indicates that the use of a low-molecular-weight contrast medium and a macromolecular contrast medium provides complementary information and suggests that the use of two contrast media within the same MR imaging session is feasible.


Subject(s)
Benzenesulfonates/pharmacology , Blood-Brain Barrier , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Contrast Media/pharmacokinetics , Glioma/drug therapy , Glioma/radiotherapy , Heterocyclic Compounds/pharmacokinetics , Magnetic Resonance Imaging/methods , Organometallic Compounds/pharmacokinetics , Pyridines/pharmacology , Analysis of Variance , Animals , Area Under Curve , Disease Models, Animal , Macromolecular Substances , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Random Allocation , Rats , Sorafenib
8.
FASEB J ; 23(9): 2986-95, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19395476

ABSTRACT

There is a need for novel strategies that target tumor vasculature, specifically those that synergize with cytotoxic therapy, in order to overcome resistance that can develop with current therapeutics. A chemistry-driven drug discovery screen was employed to identify novel compounds that inhibit endothelial cell tubule formation. Cell-based phenotypic screening revealed that noncytotoxic concentrations of (Z)-(+/-)-2-(1-benzenesulfonylindol-3-ylmethylene)-1-azabicyclo[2. 2.2]octan-3-ol (analog I) and (Z)-(+/-)-2-(1-benzylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol (analog II) inhibited endothelial cell migration and the ability to form capillary-like structures in Matrigel by > or =70%. The ability to undergo neoangiogenesis, as measured in a window-chamber model, was also inhibited by 70%. Screening of biochemical pathways revealed that analog II inhibited the enzyme ENOX1 (EC(50) = 10 microM). Retroviral-mediated shRNA suppression of endothelial ENOX1 expression inhibited cell migration and tubule formation, recapitulating the effects observed with the small-molecule analogs. Genetic or chemical suppression of ENOX1 significantly increased radiation-mediated Caspase3-activated apoptosis, coincident with suppression of p70S6K1 phosphorylation. Administration of analog II prior to fractionated X-irradiation significantly diminished the number and density of tumor microvessels, as well as delayed syngeneic and xenograft tumor growth compared to results obtained with radiation alone. Analysis of necropsies suggests that the analog was well tolerated. These results suggest that targeting ENOX1 activity represents a novel therapeutic strategy for enhancing the radiation response of tumors.


Subject(s)
Endothelium, Vascular/cytology , Neovascularization, Pathologic/drug therapy , Protein Disulfide Reductase (Glutathione)/antagonists & inhibitors , Quinuclidines/pharmacology , Transcription Factors/antagonists & inhibitors , Cell Movement/drug effects , Cell Shape/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Indoles , Membrane Proteins/antagonists & inhibitors , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/radiotherapy , Quinuclidines/therapeutic use
9.
Invest New Drugs ; 26(1): 13-24, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17805485

ABSTRACT

Radiation therapy is a widely used treatment for metastatic bone cancer, but the rapid onset of tumor radioresistance is a major problem. We investigated the radiosensitizing effect of enzastaurin, a protein kinase Cbeta (PKCbeta) inhibitor, on bone tumor growth and tumor-related pain. We found that enzastaurin enhanced the effect of ionizing radiation on cultured murine 4T1 breast cancer and murine endothelial cells, suppressing their proliferation and colony formation. Enzastaurin and ionizing radiation also induced caspase-mediated apoptosis of 4T1 cells to a greater degree than radiation alone. Enzastaurin treatment of 4T1 cells blocked the phosphorylation of PKCbeta, as well as Ras and two of its downstream effectors ERK1/2 and RAL-GTP. Using an orthotopic model of bone metastasis, we observed that a combination of enzastaurin and localized radiation treatment reduced tumor blood vessel density, bone destruction and pain compared to single modality treatment. In conclusion, we demonstrate that inhibition of PKCbeta in combination with localized radiation treatment suppresses tumor growth and alleviates pain as compared to radiation-only treatment. We also show that the radiosensitizing effect of enzastaurin is associated with suppression of tumor cell proliferation and tumor-induced angiogenesis possibly through inhibition of the Ras pathway.


Subject(s)
Indoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Protein Kinase C/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Neoplasms/drug therapy , Bone Neoplasms/radiotherapy , Bone Neoplasms/secondary , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Female , Indoles/therapeutic use , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C3H , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/radiation effects , Pain/drug therapy , Pain/radiotherapy , Protein Kinase C beta , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy, Adjuvant/methods , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Burden/radiation effects , ras Proteins/metabolism
10.
Exp Mol Med ; 40(6): 617-28, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19116447

ABSTRACT

Honokiol is an active compound purified from magnolia that has been shown to induce cell differentiation, apoptosis, and anti-angiogenesis effects, as well as an enhancement in tumor growth delay in combination with chemotherapeutic agents in several mouse xenograft models. Our goal was to investigate the radiosensitization effect of honokiol on lung carcinoma. The radiosensitization effect of liposomal honokiol in Lewis lung carcinoma cells (LL/2) was analyzed using an in vitro clonogenic survival assay. For an in vivo study, Lewis lung carcinoma-bearing C57BL/6 mice were treated with either liposomal honokiol at 25 mg/kg or 5 Gy of single tumor radiation, or a combination of both over 12 days of treatment. The tumor growth delay and the survival time were evaluated. In addition, histological analysis of tumor sections was performed to examine changes by detecting the microvessel density and apoptosis in tumor tissues. In the clonogenic survival assay, LL/2 cells treated with IC(50) Lipo-HNK for 24 h showed a radiation enhancement ratio of 1.9. After 12 days of combination treatment, the tumor volume decreased 78% and produced an anti-tumor activity 1.3-fold greater than a predicted additive effect of honokiol and radiation alone. This combination treatment also caused an 8.7 day delay in tumor growth. The cell cycle distribution and histological analysis demonstrated that liposomal honokiol has an anti-tumor effect via inducing apoptosis and inhibiting angiogenesis. Liposomal honokiol can enhance tumor cell radiosensitivity in vitro and in vivo, indicating that radiotherapy combined with liposomal honokiol can lead to greater anti-tumor efficacy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Biphenyl Compounds/therapeutic use , Carcinoma, Lewis Lung/therapy , Lignans/therapeutic use , Lung Neoplasms/therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Apoptosis , Biphenyl Compounds/administration & dosage , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Lignans/administration & dosage , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Magnolia/chemistry , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Radiation Tolerance , Transplantation, Heterologous
11.
Oral Oncol ; 42(1): 66-72, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16140034

ABSTRACT

Blocking angiogenesis may enhance conventional anticancer treatments such as radiation therapy. In this study, we examined the effects of the angiogenesis inhibitor TNP-470 on human OSCC cell lines HSC2 and KB, with combining radiation therapy in the nude mouse. We evaluated cell-induced neovascularization with dorsal air sac assay, and selected two cells (HSC2: low, KB: high) with different level of cell-induced angiogenesis. The angiogenesis inhibitor TNP-470 was given 30 mg/kg s.c. daily on day 1-5, and irradiation, 8 Gy x 1, was administered on day 1 each week for 3 weeks. Significant inhibition of tumor growth relative to untreated controls was achieved in KB cells showing high induced angiogenesis with both TNP-470 (P < 0.01) and radiation (P < 0.01) and combining TNP-470 and radiation (P < 0.01). We saw little effect of TNP-470 either alone or in addition to the effect of radiation on the HSC2 cells showing low induced angiogenesis. These results suggested that TNP-470 significantly enhanced the effect of radiation on the cells with high neovascularization. These findings indicated that individual evaluation of each tumor neovascularization potential will be important before deciding the anti-angiogenesis treatment.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/radiotherapy , Mouth Neoplasms/radiotherapy , Neovascularization, Pathologic , Sesquiterpenes/therapeutic use , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Combined Modality Therapy , Cyclohexanes , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/blood supply , Mouth Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , O-(Chloroacetylcarbamoyl)fumagillol , Tumor Cells, Cultured
12.
Photodermatol Photoimmunol Photomed ; 20(4): 191-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15238097

ABSTRACT

BACKGROUND: Photochemotherapy has long been used in the treatment of psoriasis; however, its mechanism has not been completely elucidated. Psoriasis is now regarded as an angiogenesis-related disease. Recent studies indicated that the inhibition of angiogenesis by photochemotherapy could be an underlying mechanism. It was found that photochemotherapy can downregulate the expression of angiogenic factors in keratinocytes. However, the direct effect of photochemotherapy on endothelial cells has not been studied. METHODS: In this study, we determined the effect of photochemotherapy on the proliferation of human microvascular endothelial cells through MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and cell cycle analysis. The migration assay and in vitro tube formation assay were used to investigate the migration properties and tube formation ability of human microvascular endothelial cells after psoralen plus UVA (PUVA) treatment. The apoptosis of endothelial cells elicited by photochemotherapy was also analyzed with fluorescence-activated cell sorting analysis (FACS). RESULTS: UVA (0.8-5.0 J/cm(2)) irradiation with the presence of 8-methoxypsoralen (8-MOP) (300 ng/ml) resulted in a dose-dependent reduction in the cell viabilities of endothelial cells. FACS data showed an accumulation of cells in G0/G1 phase of cell cycle and apoptotic features of cell death after UVA irradiation with psoralen. The migration properties and tube formation ability of endothelial cells were dramatically inhibited by photochemotherapy. CONCLUSION: Our results showed that photochemotherapy inhibits angiogenesis and induces apoptosis of human microvascular endothelial cells in vitro, which may be a possible mechanism of photochemotherapy in the treatment of psoriasis.


Subject(s)
Apoptosis/drug effects , Endothelium/cytology , Neovascularization, Pathologic/drug therapy , PUVA Therapy , Analysis of Variance , Apoptosis/radiation effects , Cell Movement/drug effects , Cell Movement/radiation effects , Cells, Cultured , Humans , In Vitro Techniques , Neovascularization, Pathologic/radiotherapy
13.
J Natl Cancer Inst ; 95(19): 1440-52, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14519750

ABSTRACT

Results of preclinical studies suggesting that the efficacy of molecular therapies is enhanced when they are combined with radiation have generated a surge of clinical trials combining these modalities. We reviewed the literature to identify the rationale and experimental foundation supporting the use of cyclooxygenase-2 (COX-2) inhibitors with standard radiotherapy regimens in current clinical trials. Radiation affects the ability of cells to divide and proliferate and induces the expression of genes involved in signaling pathways that promote cell survival or trigger cell death. Future advances in radiotherapy will hinge on understanding mechanisms by which radiation-induced transcription of genes governs cell death and survival, the selective control of this process, and the optimal approaches to combining this knowledge with existing therapeutic modalities. COX-2 is expressed in all stages of cancer, and in several cancers its overexpression is associated with poor prognosis. Evidence from clinical and preclinical studies indicates that COX-2-derived prostaglandins participate in carcinogenesis, inflammation, immune response suppression, apoptosis inhibition, angiogenesis, and tumor cell invasion and metastasis. Clinical trial results have demonstrated that selective inhibition of COX-2 can alter the development and the progression of cancer. In animal models, selective inhibition of COX-2 activity is associated with the enhanced radiation sensitivity of tumors without appreciably increasing the effects of radiation on normal tissue, and preclinical evidence suggests that the principal mechanism of radiation potentiation through selective COX-2 inhibition is the direct increase in cellular radiation sensitivity and the direct inhibition of tumor neovascularization. Results of current early-phase studies of non-small-cell lung, esophageal, cervical, and brain cancers will determine whether therapies that combine COX-2 inhibitors and radiation will enter randomized clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/radiotherapy , Prostaglandins/biosynthesis , Radiation-Sensitizing Agents/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Celecoxib , Chemotherapy, Adjuvant , Clinical Trials as Topic , Cyclooxygenase 2 , Disease Progression , Endothelial Growth Factors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Membrane Proteins , Neoplasms/blood supply , Neoplasms/enzymology , Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Prostaglandin-Endoperoxide Synthases , Prostaglandins/genetics , Prostaglandins/radiation effects , Pyrazoles , Radiotherapy, Adjuvant , Sulfonamides/therapeutic use , Survival Analysis , Transcription, Genetic/radiation effects , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Surg Res ; 97(2): 131-7, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11341788

ABSTRACT

INTRODUCTION: Expression of somatostatin receptor subtype 2 (sst 2) in angiogenic tumor vessels appears to be homogeneous, while tumor cell expression of this receptor is often heterogeneous. We have developed a novel in vitro three-dimensional tumor angiogenesis model to study the antitumor and the antiangiogenic effects of radiolabeled somatostatin analogs. We hypothesized that targeted in situ radiation with an Auger electron-emitting radiolabeled somatostatin analog would produce receptor-specific cytotoxicity in sst 2-expressing cells. MATERIALS AND METHODS: IMR-32 human neuroblastoma (sst 2-positive) and MDA MB-231 human breast cancer (sst 2-negative) xenografts were created in nude mice from monolayer cell cultures. Fragments of these tumors were embedded in three-dimensional fibrin gels supplemented with endothelial growth media and incubated for a period of 14 days. Tumor fragments were treated with 50 microCi/ml of (111)In-JIC 2DL, a sst 2-preferring somatostatin analog, or medium on Day 1. Initial angiogenic activity was determined at 48 h and the mean angiogenic score and tumoricidal responses were assessed on Day 14. RESULTS AND CONCLUSION: Tumoricidal effects of (111)In-JIC 2DL were seen only in sst 2-positive IMR-32 tumors. However, the angiogenic response was inhibited in both IMR-32 and MDA MB-231 tumors independent of the tumor cells' sst 2 status. Somatostatin receptor-mediated in situ radiation therapy has profound cytotoxic effects on angiogenic blood vessels and sst 2-expressing tumor cells.


Subject(s)
Contrast Media/pharmacology , Indium Radioisotopes/pharmacology , Neovascularization, Pathologic/radiotherapy , Pentetic Acid/pharmacology , Receptors, Somatostatin/metabolism , Adenocarcinoma , Amino Acid Sequence , Animals , Breast Neoplasms , Female , Humans , In Vitro Techniques , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Neuroblastoma , Octreotide/chemistry , Octreotide/pharmacology , Pentetic Acid/analogs & derivatives , Tumor Cells, Cultured
15.
Clin Cancer Res ; 6(3): 971-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741723

ABSTRACT

We examined the effect on tumor growth, vessel morphology, and expression of angiogenic factors of combining radiotherapy and antiangiogenesis in the human glioblastoma line U87 grown in the flank or intracranially in the nude mouse. The antiangiogenic agent TNP-470 was given 6.7 mg/kg s.c. daily on day 1-7 starting 1 week after transplantation. Irradiation (IR), 10 Gy x 1, was administered on day 7. A series of tumors were excised 8 and 48 h after the end of treatment. The vascular morphology was evaluated in CD31 immunostained cryosections and by electron microscopy, and the pattern of expression of angiogenic factors (mRNA and protein) was quantitatively analyzed by phosphorimaging of Northern blots and Western blots. Significant inhibition of s.c. flank tumor growth relative to untreated controls was achieved by monotherapy with both TNP-470 (P < 0.001) and IR (P < 0.001). A significant enhancement of this effect was obtained by combining TNP-470 and IR (P < 0.05). We saw no effect of TNP-470 either alone or in addition to the effect of IR on the survival of mice with intracranial tumors. CD31 immunostaining of s.c. tumors showed acute endothelial swelling and luminal protrusion in irradiated tumor vessels but never in tumors pretreated with TNP-470, and not in the untreated controls. The vessel density (Chalkley point counts) was unchanged by TNP-470 therapy. In the TNP-470-treated tumors, we observed a distinct broadening of the endothelial basement membrane by an approximately 400-700-nm-thick electron-dense yet uncharacterized fibrillar material. TNP-470 treated tumors +/- IR also had a significantly increased mRNA expression of angiopoietin-1, whereas angiopoietin-2, vascular endothelial growth factor and basic fibroblast growth factor mRNA were unchanged by the treatments. In conclusion, TNP-470 significantly enhanced the tumor effect of ionizing IR, and our findings strongly indicate that acute microvascular damage after IR is effectively prevented by concurrent TNP-470 treatment. A significant up-regulation of angiopoietin-1 seems to play a role in this protective mechanism, which as yet is not fully elucidated.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Glioblastoma/radiotherapy , Sesquiterpenes/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-1 , Angiopoietin-2 , Animals , Cell Division/drug effects , Cell Division/radiation effects , Combined Modality Therapy , Cyclohexanes , Endothelial Growth Factors/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Endothelium, Vascular/ultrastructure , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/drug therapy , Glioblastoma/ultrastructure , Humans , Lymphokines/genetics , Male , Membrane Glycoproteins/genetics , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , O-(Chloroacetylcarbamoyl)fumagillol , Proteins/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , RNA, Messenger/radiation effects , Survival Analysis , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Invest New Drugs ; 15(1): 39-48, 1997.
Article in English | MEDLINE | ID: mdl-9195288

ABSTRACT

TNP-740, minocycline, suramin and genistein have demonstrated antiangiogenic activity in various experimental systems. The effect of these agents alone and in two agent combinations on the number of intratumoral vessels and response to cytotoxic anticancer therapies was assessed in animals bearing the Lewis lung carcinoma. Treatment with each of the antiangiogenic agents alone and in two agent combinations decreased the number of intratumoral vessels visualized by CD31 or Factor VIII staining to 30% to 50% of the number in the untreated control tumors. In general, the antiangiogenic agents are more effective adjuvants to cytotoxic therapies when used as two agent combinations than as single agents. The most effective antiangiogenic combinations were: TNP-470/minocycline > TNP-470/genistein > TNP-470/suramin. The increases in the response of the primary tumor to cyclophosphamide, adriamycin, CDDP, BCNU, x-rays or 5-fluorouracil and the lung metastases occur to about the same level with the addition of antiangiogenic agents to the therapies. With the treatment combination TNP-470/minocycline/cyclophosphamide 40% of the animals were cured. The results of these studies indicate that antiangiogenic agents can be very useful additions to treatment regimens for solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Carmustine/pharmacology , Carmustine/therapeutic use , Cell Division/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cyclohexanes , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Synergism , Genistein , Isoflavones/pharmacology , Isoflavones/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Minocycline/pharmacology , Minocycline/therapeutic use , Neoplasm Invasiveness , Neovascularization, Pathologic/radiotherapy , O-(Chloroacetylcarbamoyl)fumagillol , Random Allocation , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Suramin/pharmacology , Suramin/therapeutic use
17.
Breast Cancer Res Treat ; 36(2): 227-36, 1995.
Article in English | MEDLINE | ID: mdl-8534870

ABSTRACT

The ability of the antiangiogenic agents TNP-470 and minocycline, singly or in combination, to potentiate the antitumor effects of several cytotoxic therapies was assessed in the murine EMT-6 mammary carcinoma as well as in two drug resistant sublines of that tumor designated EMT-6/CTX and EMT-6/CDDP. The antiangiogenic agents alone or in combination did not alter the growth of the tumors. However, their administration along with cyclophosphamide, CDDP, or thiotepa substantially increased the tumor growth delay produced by these cytotoxic therapies in tumors responsive to the drugs--the increase was about 2-fold for TNP-470 and minocycline together. In drug resistant tumors, treatment with the antiangiogenic agents did not reverse drug resistance but did increase the effect of the cytotoxic drugs. Treatment with TNP-470/minocycline also increased the oxygenation of each of the three tumors. Thus, TNP-470/minocycline administration increased the efficacy of fractionated radiation therapy, especially when used along with a perflubron emulsion oxygen delivery agent/carbogen. These results indicate that treatment regimens including therapies directed toward the proliferating normal cells within a tumor mass as well as therapies directed toward the malignant cells can produce improved outcomes.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mammary Neoplasms, Experimental/blood supply , Minocycline/pharmacology , Neovascularization, Pathologic/drug therapy , Sesquiterpenes/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Cell Division/drug effects , Combined Modality Therapy , Cyclohexanes , Drug Resistance, Neoplasm , Drug Synergism , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Minocycline/administration & dosage , Neoplasm Transplantation , Neovascularization, Pathologic/radiotherapy , O-(Chloroacetylcarbamoyl)fumagillol , Sesquiterpenes/administration & dosage , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL