Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 622
Filter
2.
Mol Carcinog ; 60(7): 481-496, 2021 07.
Article in English | MEDLINE | ID: mdl-34018249

ABSTRACT

c-Hepatocyte growth factor receptor (Met) inhibitors have demonstrated clinical benefits in some types of solid tumors. However, the efficacy of c-Met inhibitors in esophageal squamous cell carcinoma (ESCC) remains unclear. In this study, we discovered that c-Met inhibitors induced "Signal Transducer and Activator of Transcription (STAT3)-addiction" in ESCC cells, and the feedback activation of STAT3 in ESCC cells limits the tumor response to c-Met inhibition. Mechanistically, c-Met inhibition increased the autocrine of several cytokines, including CCL2, interleukin 8, or leukemia inhibitory factor, and facilitated the interactions between the receptors of these cytokines and Janus Kinase1/2 (JAK1/2) to resultantly activate JAKs/STAT3 signaling. Pharmacological inhibition of c-Met together with cytokines/JAKs/STAT3 axis enhanced cancer cells regression in vitro. Importantly, combined c-Met and STAT3 inhibitors synergistically suppressed tumor growth and promoted the apoptosis of tumor cells without producing systematic toxicity. These findings suggest that inhibition of the STAT3 feedback loop may augment the response to c-Met inhibitors via the STAT3-mediated oncogene addiction in ESCC cells.


Subject(s)
Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Aminosalicylic Acids/administration & dosage , Aminosalicylic Acids/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacology , Drug Resistance, Neoplasm , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/mortality , Feedback, Physiological/drug effects , Female , Humans , Male , Mice, Inbred BALB C , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Tyrosine/metabolism , Xenograft Model Antitumor Assays
3.
BMC Cancer ; 21(1): 270, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33711962

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is a target for cancer therapy as it is overexpressed in a wide variety of cancers. Therapeutic antibodies that bind EGFR are being evaluated in clinical trials as imaging agents for positron emission tomography and image-guided surgery. However, some of these antibodies have safety concerns such as infusion reactions, limiting their use in imaging applications. Nimotuzumab is a therapeutic monoclonal antibody that is specific for EGFR and has been used as a therapy in a number of countries. METHODS: Formulation of IRDye800CW-nimotuzumab for a clinical trial application was prepared. The physical, chemical, and pharmaceutical properties were tested to develop the specifications to determine stability of the product. The acute and delayed toxicities were tested and IRDye800CW-nimotuzumab was determined to be non-toxic. Non-compartmental pharmacokinetics analysis was used to determine the half-life of IRDye800CW-nimotuzumab. RESULTS: IRDye800CW-nimotuzumab was determined to be non-toxic from the acute and delayed toxicity study. The half-life of IRDye800CW-nimotuzumab was determined to be 38 ± 1.5 h. A bi-exponential analysis was also used which gave a t1/2 alpha of 1.5 h and t1/2 beta of 40.8 h. CONCLUSIONS: Here, we show preclinical studies demonstrating that nimotuzumab conjugated to IRDye800CW is safe and does not exhibit toxicities commonly associated with EGFR targeting antibodies.


Subject(s)
Drugs, Investigational/administration & dosage , Immunoconjugates/administration & dosage , Neoplasms/diagnostic imaging , Optical Imaging/methods , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/toxicity , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacokinetics , Benzenesulfonates/toxicity , Cell Line, Tumor , Clinical Trials as Topic , Drug Stability , Drugs, Investigational/pharmacology , Drugs, Investigational/toxicity , ErbB Receptors/antagonists & inhibitors , Female , Half-Life , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/toxicity , Investigational New Drug Application , Male , Mice , Neoplasms/pathology , Neoplasms/surgery , Surgery, Computer-Assisted/methods , Toxicity Tests, Acute , Xenograft Model Antitumor Assays
4.
J Surg Res ; 233: 20-25, 2019 01.
Article in English | MEDLINE | ID: mdl-30502249

ABSTRACT

BACKGROUND: Loss of intestinal barrier integrity plays a fundamental role in the pathogenesis of various gastrointestinal diseases and is implicated in the onset of sepsis and multiple organ failure. An array of methods to assess different aspects of intestinal barrier function suffers from lack of sensitivity, prolonged periods of specimen collection, or high expense. We have developed a technique to measure the concentration of the food dye FD&C Blue #1 from blood and sought to assess its utility in measuring intestinal barrier function in humans. MATERIALS AND METHODS: Four healthy volunteers and 10 critically ill subjects in the intensive care unit were recruited in accordance with an institutional review board approved protocol. Subjects were given 0.5 mg/kg Blue #1 enterally as an aqueous solution of diluted food coloring. Five blood specimens were drawn per subject: 0 h (before dose), 1, 2, 4, and 8 h. After plasma isolation, organic extracts were analyzed by high-performance liquid chromatography/mass spectrometry detecting the presence of unmodified dye. RESULTS: We found no baseline detectable absorption in healthy volunteers. After including the subjects in the intensive care unit, we compared dye absorption in the six subjects who met criteria for septic shock with the eight who did not. Septic patients demonstrated significantly greater absorption of Blue #1 after 2 h. CONCLUSIONS: We have developed a novel, easy-to-use method to measure intestinal barrier integrity using a food grade dye detectable by mass spectrometry analysis of patient blood following oral administration.


Subject(s)
Food Coloring Agents/pharmacokinetics , Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Shock, Septic/diagnosis , Administration, Oral , Adult , Benzenesulfonates/administration & dosage , Benzenesulfonates/blood , Benzenesulfonates/pharmacokinetics , Critical Illness , Feasibility Studies , Female , Food Coloring Agents/administration & dosage , Food Coloring Agents/analysis , Healthy Volunteers , Humans , Intensive Care Units , Male , Permeability , Prospective Studies , Shock, Septic/blood , Shock, Septic/physiopathology
5.
J Surg Res ; 239: 44-51, 2019 07.
Article in English | MEDLINE | ID: mdl-30798171

ABSTRACT

BACKGROUND: Fluorescence-guided surgery (FGS) is a rapidly advancing field that may improve outcomes in several cancer types. Although screening has decreased colorectal cancer (CRC) mortality, it remains a common and often fatal malignancy. In this study, we sought to identify an optical imaging agent for the application of FGS technology to CRC. METHODS: We compared a panitumumab-IRDye800CW conjugate to an IgG-IRDye800CW isotype control. Mice were implanted with one of three CRC cell lines (LS174T, Colo205, and SW948) and imaged with open- and closed-filed fluorescence imaging systems. Fluorescent contrast was quantified by calculating the ratio between tumor and background fluorescence. After 10 d, the mice were sacrificed, and their tumors stained for microscopic imaging. RESULTS: Panitumumab-IRDye800CW produced significantly greater (P < 0.05) fluorescent contrast in all three cell lines. Average tumor to background ratio was 6.00 versus 2.60 for LS174T, 5.78 versus 2.52 for Colo205, and 4.31 versus 1.70 for SW948. A 1-mg tumor fragment produced significantly greater fluorescent contrast in the Colo205 and SW948 cell lines in the panitumumab-IRDye800CW group. Western blotting for epidermal growth factor receptor (EGFR) and a semiquantitative analysis of EGFR expression noted strong expression in all three cell lines; however, EGFR expression did not directly correlate to tumor to background ratio. CONCLUSIONS: Panitumumab-IRDye800CW produces significantly greater fluorescent contrast than IgG-IRDye800CW in a murine model of CRC and is a suitable agent for the application of FGS technology to CRC.


Subject(s)
Benzenesulfonates/administration & dosage , Colorectal Neoplasms/surgery , Immunoconjugates/administration & dosage , Indoles/administration & dosage , Optical Imaging/methods , Panitumumab/administration & dosage , Surgery, Computer-Assisted/methods , Animals , Cell Line, Tumor , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Humans , Image Processing, Computer-Assisted , Immunoconjugates/chemistry , Injections, Intravenous , Mice , Mice, Nude , Optical Imaging/instrumentation , Surgery, Computer-Assisted/instrumentation , Xenograft Model Antitumor Assays
6.
J Surg Res ; 236: 172-183, 2019 04.
Article in English | MEDLINE | ID: mdl-30694753

ABSTRACT

BACKGROUND: Acute liver failure (ALF) from severe acute liver injury is a critical condition associated with high mortality. The purpose of this study was to investigate the impact of preemptive administration of γ-aminobutyric acid (GABA) on hepatic injury and survival outcomes in mice with experimentally induced ALF. MATERIALS AND METHODS: To induce ALF, C57BL/6NHsd mice were administered GABA, saline, or nothing for 7 d, followed by intraperitoneal administration of 500 µg of tumor necrosis factor α and 20 mg of D-galactosamine. The study mice were humanely euthanized 4-5 h after ALF was induced or observed for survival. Proteins present in the blood samples and liver tissue from the euthanized mice were analyzed using Western blot and immunohistochemical and histopathologic analyses. For inhibition studies, we administered the STAT3-specific inhibitor, NSC74859, 90 min before ALF induction. RESULTS: We found that GABA-treated mice had substantial attenuation of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive hepatocytes and hepatocellular necrosis, decreased caspase-3, H2AX, and p38 MAPK protein levels and increased expressions of Jak2, STAT3, Bcl-2, and Mn-SOD, with improved mitochondrial integrity. The reduced apoptotic proteins led to a significantly prolonged survival after ALF induction in GABA-treated mice. The STAT3-specific inhibitor NSC74859 eliminated the survival advantage in GABA-treated mice with ALF, indicating the involvement of the STAT3 pathway in GABA-induced reduction in apoptosis. CONCLUSIONS: Our results showed that preemptive treatment with GABA protected against severe acute liver injury in mice via GABA-mediated STAT3 signaling. Preemptive administration of GABA may be a useful approach to optimize marginal donor livers before transplantation.


Subject(s)
Liver Failure, Acute/prevention & control , Liver/drug effects , Protective Agents/administration & dosage , gamma-Aminobutyric Acid/administration & dosage , Aminosalicylic Acids/administration & dosage , Animals , Benzenesulfonates/administration & dosage , Disease Models, Animal , Galactosamine/toxicity , Humans , Injections, Intraperitoneal , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Necrosis/chemically induced , Necrosis/pathology , Necrosis/prevention & control , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Treatment Outcome , Tumor Necrosis Factor-alpha/toxicity
7.
J Surg Res ; 242: 145-150, 2019 10.
Article in English | MEDLINE | ID: mdl-31077946

ABSTRACT

INTRODUCTION: Claudins are tight-junction proteins, which maintain an epithelial barrier in normal colon cells. Overexpression of Claudin-1 has been implicated for development of colon cancer. We postulated that Claudin-1 may be a useful target in near-infrared imaging and fluorescence-guided surgery. METHODS: We conjugated Claudin-1 antibody to LI-COR IR800DyeCW (Claudin-1-IRDye800CW). Western blotting of 9 human colon cancer cell lysates was performed. Animal imaging was performed with the LI-COR Pearl Trilogy Fluorescence Imaging System. A dose-response study was carried out with subcutaneous LS174T colon cancer cell line models. Increasing doses of Claudin-1-IRDye800CW via tail vein injection were administered to three groups of mice. Two groups of mice were used as controls (antibody alone, and dye alone). In vivo imaging was performed at 24, 48, and 72 h after administration of the conjugated dye. Orthotopic implantation of patient-derived tumors and cell lines was performed and peritoneal carcinomatosis models were created. After tumor growth, mice were administered Claudin-1-IRDye800CW and imaged in vivo 48 h later. The mice were euthanized and laparotomy was performed to assess internal organs and toxicity. RESULTS: Western blotting revealed that all colon cancer cell lysates expressed varying amounts of Claudin-1. All tumors demonstrated strong and specific fluorescence labeling at 800 nm, even with the lowest dose of 12.5 µg of Claudin-1-IRDye800CW. CONCLUSIONS: Claudin-1 is a useful target for near-infrared antibody-based imaging for visualization of colorectal tumors for future use in fluorescence-guided surgery.


Subject(s)
Claudin-1/immunology , Colonic Neoplasms/diagnostic imaging , Fluorescent Dyes/administration & dosage , Immunoconjugates/administration & dosage , Peritoneal Neoplasms/diagnostic imaging , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Benzenesulfonates/administration & dosage , Cell Line, Tumor , Colon/diagnostic imaging , Colon/pathology , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Dose-Response Relationship, Drug , Humans , Immunoconjugates/immunology , Indoles/administration & dosage , Injections, Intravenous , Male , Mice , Mice, Nude , Peritoneal Neoplasms/surgery , Spectroscopy, Near-Infrared/methods , Surgery, Computer-Assisted/methods , Xenograft Model Antitumor Assays
8.
J Pharmacol Sci ; 139(3): 180-185, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30738725

ABSTRACT

We analyzed how the enhancement of net sarcoplasmic reticulum (SR) Ca2+ uptake may affect cardiac electrophysiological properties in vivo by using caldaret which can decrease SR diastolic Ca2+ leak, enhance SR Ca2+ reuptake and inhibit reverse-mode Na+/Ca2+ exchanger. Caldaret in doses of 0.5, 5 and 50 µg/kg was intravenously administered over 10 min to the halothane-anesthetized beagle dogs (n = 5), attaining pharmacologically active plasma concentration. The low and middle doses of caldaret increased the ventricular contraction, which could be explained by its on-target pharmacological activities. The high dose enhanced the sinus automaticity followed by its suppression in addition to the increase of the total peripheral resistance, which may be unfavorable for treating diastolic heart failure. The low and middle doses enhanced the atrioventricular conduction, which may have some potential for predisposing the atria to the onset of atrial fibrillation via an induction of mitral and/or tricuspid regurgitation. The middle and high doses of caldaret prolonged the ventricular effective refractory period without altering the intraventricular conduction or repolarization period, which may prevent the onset of ventricular arrhythmias. Thus, modulation of intracellular Ca2+ handling by caldaret can induce not only inotropic effect, but also various electrophysiological actions on the in situ heart.


Subject(s)
Benzenesulfonates/pharmacology , Calcium/administration & dosage , Cardiotonic Agents/pharmacology , Piperazines/pharmacology , Sarcoplasmic Reticulum/drug effects , Animals , Arrhythmias, Cardiac/prevention & control , Benzenesulfonates/administration & dosage , Calcium/metabolism , Cardiotonic Agents/administration & dosage , Dogs , Dose-Response Relationship, Drug , Electrophysiologic Techniques, Cardiac , Female , Halothane/administration & dosage , Heart Atria/drug effects , Heart Atria/metabolism , Piperazines/administration & dosage , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism
9.
J Pharm Pharm Sci ; 21(1s): 325s-334s, 2018.
Article in English | MEDLINE | ID: mdl-30465708

ABSTRACT

Interleukin (IL)-6 decreases hepatic expression of numerous transporters. Although IL-6 signaling occurs through STAT3, the extent of the involvement of the STAT3 signaling pathway has not been elucidated. PURPOSE: Our objective was to investigate whether IL-6-mediated effects occur through STAT3, and whether PXR plays a role in this regulation. METHOD: PXR null (-/-) or wild-type (+/+) male mice were pre-dosed with a selective STAT3 inhibitor S3I-201 (7.5 mg/kg ip) or vehicle (n=5-8/group) 30 minutes before receiving a single dose of IL-6 (1 µg ip) or saline. Animals were sacrificed after 6 hours and liver samples were analyzed using qRT-PCR and western blotting. RESULTS: As compared to saline controls, IL-6 decreased the expression of Cyp3a, Abcb1a, Abcc3, and Slco1a4 20-70% similarly in PXR (+/+) and (-/-) mice at 6 hr, while downregulation of Abcb11, Abcc2, Slc10a1and Slco2b1 was only seen in PXR (+/+). Pre-administration of S3I-201 attenuated IL-6-mediated changes of most transporters in PXR (+/+) and PXR (-/-) mice. At early times after IL-6 administration (10-120 minutes), transcript levels of Socs3, PXR, Abcb1a, Abcc3, Abcb11, Slco1a4 and Slco2b1were increased in PXR (+/+) mice. CONCLUSIONS: Our findings demonstrate that IL-6 imposes a significant downregulation of numerous ABC and SLC transporters in liver primarily through activation of the STAT3 signaling pathway. Based on time-dependent changes in transporter expression, downregulation likely occurs downstream of STAT3 activation.  As IL-6 is elevated in many diseases, understanding the underlying mechanism(s) involved in transporter dysregulation will allow us to predict potential drug-disease interactions.


Subject(s)
Down-Regulation , Interleukin-6/metabolism , Pregnane X Receptor/metabolism , STAT3 Transcription Factor/metabolism , Aminosalicylic Acids/administration & dosage , Aminosalicylic Acids/pharmacology , Animals , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacology , Down-Regulation/drug effects , Injections, Intraperitoneal , Interleukin-6/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnane X Receptor/deficiency , Pregnane X Receptor/genetics , STAT3 Transcription Factor/antagonists & inhibitors
10.
Mol Pharm ; 14(4): 1145-1153, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28245129

ABSTRACT

Advances in optical imaging technologies have stimulated the development of near-infrared (NIR) fluorescently labeled targeted probes for use in image-guided surgery. As nanobodies have already proven to be excellent candidates for molecular imaging, we aimed in this project to design NIR-conjugated nanobodies targeting the tumor biomarker HER2 for future applications in this field and to evaluate the effect of dye and dye conjugation chemistry on their pharmacokinetics during development. IRDye800CW or IRdye680RD were conjugated either randomly (via lysines) or site-specifically (via C-terminal cysteine) to the anti-HER2 nanobody 2Rs15d. After verification of purity and functionality, the biodistribution and tumor targeting of the NIR-nanobodies were assessed in HER2-positive and -negative xenografted mice. Site-specifically IRDye800CW- and IRdye680RD-labeled 2Rs15d as well as randomly labeled 2Rs15d-IRDye680RD showed rapid tumor accumulation and low nonspecific uptake, resulting in high tumor-to-muscle ratios at early time points (respectively 6.6 ± 1.0, 3.4 ± 1.6, and 3.5 ± 0.9 for HER2-postive tumors at 3 h p.i., while <1.0 for HER2-negative tumors at 3 h p.i., p < 0.05). Contrarily, using the randomly labeled 2Rs15d-IRDye800CW, HER2-positive and -negative tumors could only be distinguished after 24 h due to high nonspecific signals. Moreover, both randomly labeled 2Rs15d nanobodies were not only cleared via the kidneys but also partially via the hepatobiliary route. In conclusion, near-infrared fluorescent labeling of nanobodies allows rapid, specific, and high contrast in vivo tumor imaging. Nevertheless, the fluorescent dye as well as the chosen conjugation strategy can affect the nanobodies' properties and consequently have a major impact on their pharmacokinetics.


Subject(s)
Benzenesulfonates/administration & dosage , Indoles/administration & dosage , Nanoparticles/metabolism , Neoplasms/diagnosis , Single-Domain Antibodies/metabolism , Tissue Distribution/drug effects , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetulus , Female , Mice , Mice, Nude , Molecular Imaging/methods , Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Spectroscopy, Near-Infrared/methods , Surgery, Computer-Assisted/methods
11.
Mol Pharm ; 14(10): 3457-3463, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28826214

ABSTRACT

Complete resection of tumor lesions in advanced stage ovarian cancer patients is of utmost importance, since the extent of residual disease after surgery strongly affects survival. Intraoperative imaging may be useful to improve surgery in these patients. Farletuzumab is a humanized IgG1 antibody that specifically recognizes the folate receptor alpha (FRα). Labeled with a radiolabel and a fluorescent dye, farletuzumab may be used for the intraoperative detection of ovarian cancer lesions. The current aim is to demonstrate the feasibility of FRα-targeted dual-modality imaging using 111In-farletuzumab-IRDye800CW in an intraperitoneal ovarian cancer model. Biodistribution studies were performed 3 days after injection of 3, 10, 30, or 100 µg of 111In-farletuzumab-IRDye800CW in mice with subcutaneous IGROV-1 tumors (5 mice per group). In mice with intraperitoneal IGROV-1 tumors the nonspecific uptake of 111In-farletuzumab-IRDye800CW was determined by coinjecting an excess of unlabeled farletuzumab. MicroSPECT/CT and fluorescence imaging were performed 3 days after injection of 10 µg of 111In-farletuzumab-IRDye800CW. FRα expression in tumors was determined immunohistochemically. Optimal tumor-to-blood-ratios (3.4-3.7) were obtained at protein doses up to 30 µg. Multiple intra-abdominal tumor lesions were clearly visualized by microSPECT/CT, while uptake in normal tissues was limited. Fluorescence imaging was used to visualize and guide resection of superficial tumors. Coinjection of an excess of unlabeled farletuzumab significantly decreased tumor uptake of 111In-farletuzumab-IRDye800CW (69.4 ± 27.6 versus 18.3 ± 2.2% ID/g, p < 0.05). Immunohistochemical analyses demonstrated that the radioactive and fluorescent signal corresponded with FRα-expressing tumor lesions. FRα-targeted SPECT/fluorescence imaging using 111In-farletuzumab-IRDye800CW can be used to detect ovarian cancer in vivo and could be a valuable tool for enhanced intraoperative tumor visualization in patients with intraperitoneal metastases of ovarian cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Folate Receptor 1/antagonists & inhibitors , Intraoperative Care/methods , Ovarian Neoplasms/diagnostic imaging , Surgery, Computer-Assisted/methods , Animals , Antibodies, Monoclonal, Humanized/chemistry , Benzenesulfonates/administration & dosage , Benzenesulfonates/chemistry , Cell Line, Tumor , Feasibility Studies , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Folate Receptor 1/immunology , Humans , Indium Radioisotopes/administration & dosage , Indium Radioisotopes/chemistry , Indoles/administration & dosage , Indoles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging/methods , Optical Imaging/methods , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methods , Xenograft Model Antitumor Assays
12.
J Surg Oncol ; 116(7): 898-906, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28628728

ABSTRACT

BACKGROUND AND OBJECTIVES: Optical imaging to guide cancer resections is rapidly transitioning into the operating room. However, the sensitivity of this technique to detect subclinical disease is yet characterized. The purpose of this study was to determine the minimum range of cancer cells that can be detected by antibody-based fluorescence imaging. METHODS: 2LMP (breast), COLO-205 (colon), MiaPaca-2 (pancreas), and SCC-1 (head and neck) cells incubated in vitro with cetuximab-IRDye800CW (dose range 8.6-86 nM) were implanted subcutaneously in mice (n = 3 mice, 5 tumors/mouse). Following incubation with 8.6 × 10-2 µM of cetuximab-IRDye800CW in vitro, serial dilutions of each cell type (1 × 103 -1 × 106 ) were implanted subcutaneously (n = 3, 5 tumors/mouse). Tumors were imaged with Pearl Impulse and Xenogen IVIS 100 imaging systems. Scatchard analysis was performed to determine receptor density and kinetics for each cell line. RESULTS: Under conditions of minimal cetuximab-IRDye800CW exposure to low cellular quantity, closed-field fluorescence imaging theoretically detected a minimum of 4.2 × 104 -9.5 × 104 2LMP cells, 1.9 × 105 -4.5 × 105 MiaPaca-2 cells, and 2.4 × 104 -6.7 × 104 SCC-1 cells; COLO-205 cells could not be identified. Higher EGFR-mediated uptake of cetuximab correlated with sensitivity of detection. CONCLUSION: This study supports the clinical utility of cetuximab-IRDye800CW to sensitively localize subclinical disease in the surgical setting.


Subject(s)
Neoplasms/diagnostic imaging , Neoplasms/surgery , Optical Imaging/methods , Surgery, Computer-Assisted/methods , Animals , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacokinetics , Cell Line, Tumor , Cetuximab/administration & dosage , Cetuximab/pharmacokinetics , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Heterografts , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Mice , Mice, Nude , Organotechnetium Compounds/administration & dosage , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics
13.
Retina ; 36(3): 558-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26352556

ABSTRACT

PURPOSE: To describe the clinical characteristics of epiretinal membranes (ERMs) secondary to nonprogressive diseases in very young children and to assess their surgical outcome. METHODS: Retrospective interventional case series study of 13 pediatric patients (aged less than 12 years) operated on for ERM more than a 6-year period. Visual acuity measurement, ophthalmic examination, and optical coherence tomography imaging were assessed preoperatively and postoperatively. RESULTS: Mean age was 6.5 years (3-12 years). Mean follow-up duration was of 28.5 months (12-69 months). Among the 13 patients, there were 7 cases of idiopathic ERM, 4 cases of combined hamartoma of the retina and retinal pigment epithelium, and 2 cases of posttraumatic ERM. The diagnosis of ERM was fortuitous in 10 cases (77%). Twelve children (92%) experienced a functional improvement. All patients received adjuvant treatment of amblyopia. Best-corrected visual acuity improved significantly after surgery from 20/160 to 20/40 (P = 0.001). CONCLUSION: Even in very young children, surgery resulted in a significant long-term improvement. Children screening was essential for diagnosis and treatment in most cases.


Subject(s)
Epiretinal Membrane/surgery , Vitrectomy , Benzenesulfonates/administration & dosage , Child , Child, Preschool , Coloring Agents/administration & dosage , Epiretinal Membrane/diagnosis , Epiretinal Membrane/physiopathology , Female , Follow-Up Studies , Humans , Male , Retrospective Studies , Tomography, Optical Coherence , Visual Acuity/physiology
14.
Retina ; 36(8): 1463-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26756806

ABSTRACT

PURPOSE: To compare the structural and functional outcome of use of autologous heparinized whole blood before staining internal limiting membrane with brilliant blue (BB) versus conventional BB-assisted macular hole surgery. METHODS: Sixty eyes of 60 patients were randomly divided equally in Group A (BB staining using whole blood) and Group B (conventional BB staining). Clinical assessment and spectral domain optical coherence tomography was done at baseline and 3 weeks, 6 weeks, 16 weeks, and 6 months postoperatively. RESULTS: Group A eyes had a significantly higher best-corrected visual acuity as compared with Group B postoperatively (P < 0.001, <0.001, 0.004, 0.04 at 3, 6, 16 weeks, and 6 months). Inner segment/outer segment junction continuity was noted in greater number of eyes in Group A compared with Group B (P = 0.02, 0.002, 0.003, and 0.03 at 3, 6, 16 weeks, and 6 months). Eyes in Group A had significantly higher outer foveal thickness at 3 weeks (P = 0.001) and 6 weeks (P < 0.001) compared with Group B. CONCLUSION: Use of whole blood before staining internal limiting membrane with BB causes earlier and better visual rehabilitation postoperatively, which could be attributed to earlier photoreceptor regeneration as evidenced by inner segment/outer segment junction continuity and increase in outer foveal thickness.


Subject(s)
Basement Membrane/pathology , Benzenesulfonates/administration & dosage , Blood , Coloring Agents/administration & dosage , Epiretinal Membrane/pathology , Retinal Perforations/surgery , Adult , Aged , Endotamponade , Female , Humans , Intraocular Pressure , Intraoperative Care , Male , Middle Aged , Prone Position , Retinal Perforations/physiopathology , Staining and Labeling/methods , Tomography, Optical Coherence , Visual Acuity/physiology
15.
J Transl Med ; 13: 48, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25638171

ABSTRACT

BACKGROUND: Dobesilate (2,5-dihydroxyphenyl sulfonate, DHPS) was recently identified as the most potent member of a family of fibroblast growth factor (FGF) inhibitors headed by gentisic acid, one of the main catabolites of aspirin. Although FGFs were first described as inducers of angiogenesis, they were soon recognized as broad spectrum mitogens. Furthermore, in the last decade these proteins have been shown to participate directly in the onset of inflammation, and their potential angiogenic activity often contributes to the inflammatory process in vivo. The aim of this work was to evaluate the anti-inflammatory, anti-angiogenic and anti-tumoral activities of the derivative of DHPS obtained by acetoxylation of its two hydroxyl groups (2,5-diacetoxyphenyl sulfonate; DAPS). METHODS: Anti-inflammatory, anti-angiogenic and anti-tumoral activities of DHPS and DAPS were compared using in vivo assays of dermatitis, angiogenesis and tumorigenesis. The effects of both compounds on myeloperoxidase (MPO) and cyclooxygenase (COX) activities, cytokine production and FGF-induced fibroblast proliferation were also determined. RESULTS: Topical DAPS is more effective than DHPS in preventing inflammatory signs (increased vascular permeability, edema, leukocyte infiltration, MPO activation) caused by contact dermatitis induction in rat ears. DAPS, but not DHPS, effectively inhibits COX-1 and COX-2 activities. DAPS also reduces the increase in serum cytokine concentration induced by lipopolysaccharide in rats. Furthermore, DAPS displays higher in vivo efficacy than DHPS in inhibiting FGF-induced angiogenesis and heterotopic glioma progression, with demonstrated oral efficacy to combat both processes. CONCLUSIONS: By inhibiting both FGF-signaling and COX-mediated prostaglandin synthesis, DAPS efficiently breaks the vicious circle created by the reciprocal induction of FGF and prostaglandins, which probably sustains undesirable inflammation in many circumstances. Our findings define the enhancement of anti-inflammatory, anti-angiogenic and anti-tumoral activities by diacetyloxyl derivatization of the FGF inhibitor, dobesilate.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Fibroblast Growth Factors/antagonists & inhibitors , Acetylation , Administration, Oral , Administration, Topical , Animals , Benzenesulfonates/administration & dosage , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Benzenesulfonates/therapeutic use , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dermatitis/complications , Dermatitis/drug therapy , Dermatitis/pathology , Fibroblast Growth Factors/metabolism , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Male , Mitogens/pharmacology , Rats, Sprague-Dawley
16.
BMC Cancer ; 15: 525, 2015 Jul 18.
Article in English | MEDLINE | ID: mdl-26187637

ABSTRACT

BACKGROUND: The lymph node metastasis is a key early step of the tumor metastatic process. VEGFD-mediated tumor lymphangiogenesis plays a key role, since down-regulation of p-VEGFR-3 could block the lymph node metastasis. YL529 has been reported to possess potent anti-angiogenesis and antitumor activities; however, its roles in tumor-associated lymphangiogenesis and lymphatic metastasis remain unclear. METHOD: We investigated the effect of YL529 on tumor-associated lymphangiogenesis and lymph node metastasis using in vitro lymph node metastasis models and in vivo subcutaneous tumor models in C57 BL/6 mice. RESULT: We found that YL529 inhibited VEGF-D-induced survival, proliferation and tube-formation of Human Lymphatic Endothelial Cells. Furthermore, in established in vitro and in vivo lymph node metastasis models using VEGF-D-LL/2 cells, YL529 significantly inhibited the tumor-associated lymphangiogenesis and metastasis. At molecular level, YL529 down-regulated p-VEGFR-3, p-JNK and Bax while up-regulated Bcl-2. CONCLUSION: YL529 provided the therapeutic benefits by both direct effects on tumor cells and inhibiting lymphangiogenesis and metastasis via the VEGFR-3 signaling pathway, which may have significant direct clinical implications.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Benzenesulfonates/administration & dosage , Lymphangiogenesis/drug effects , Lymphatic Metastasis/prevention & control , Picolines/administration & dosage , Vascular Endothelial Growth Factor D/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Picolines/pharmacology , Xenograft Model Antitumor Assays
17.
J Magn Reson Imaging ; 42(6): 1582-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25920494

ABSTRACT

BACKGROUND: Glioblastoma is a malignant World Health Organization (WHO) grade IV glioma with a poor prognosis in humans. New therapeutics are desperately required. The nitrone OKN-007 (2,4-disulfophenyl-PBN) has demonstrated effective anti-glioma properties in several rodent models and is currently being used as a clinical investigational drug for recurrent gliomas. We assessed the regional effects of OKN-007 in the tumor necrotic core and non-necrotic tumor parenchyma. METHODS: An F98 rat glioma model was evaluated using proton magnetic resonance spectroscopy ((1) H-MRS), diffusion-weighted imaging (DWI), morphological T2-weighted imaging (T2W) at 7 Tesla (30 cm-bore MRI), as well as immunohistochemistry and microarray assessments, at maximum tumor volumes (15-23 days following cell implantation in untreated (UT) tumors, and 18-35 days in OKN-007-treated tumors). RESULTS: (1) H-MRS data indicates that Lip0.9/Cho, Lip0.9/Cr, Lip1.3/Cho, and Lip1.3/Cr ratios are significantly decreased (all P < 0.05) in the OKN-007-treated group compared with UT F98 gliomas. The Cho/Cr ratio is also significantly decreased in the OKN-007-treated group compared with UT gliomas. In addition, the OKN-007-treated group demonstrates significantly lower ADC values in the necrotic tumor core and the nonnecrotic tumor parenchyma (both P < 0.05) compared with the UT group. There was also an increase in apoptosis following OKN-007 treatment (P < 0.01) compared with UT. CONCLUSION: OKN-007 reduces both necrosis and tumor cell proliferation, as well as seems to mediate multiple effects in different tumor regions (tumor necrotic core and nonnecrotic tumor parenchyma) in F98 gliomas, indicating the efficacy of OKN-007 as an anti-cancer agent and its potential clinical use.


Subject(s)
Benzenesulfonates/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioma/drug therapy , Glioma/pathology , Imines/administration & dosage , Magnetic Resonance Imaging/methods , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Necrosis/pathology , Necrosis/prevention & control , Rats , Rats, Inbred F344
18.
Blood ; 119(22): 5133-43, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22368270

ABSTRACT

Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 (FLT3) gene occurs in 30% of acute myeloid leukemias (AMLs) and confers a poor prognosis. Thirteen relapsed or chemo-refractory FLT3-ITD(+) AML patients were treated with sorafenib (200-400 mg twice daily). Twelve patients showed clearance or near clearance of bone marrow myeloblasts after 27 (range 21-84) days with evidence of differentiation of leukemia cells. The sorafenib response was lost in most patients after 72 (range 54-287) days but the FLT3 and downstream effectors remained suppressed. Gene expression profiling showed that leukemia cells that have become sorafenib resistant expressed several genes including ALDH1A1, JAK3, and MMP15, whose functions were unknown in AML. Nonobese diabetic/severe combined immunodeficiency mice transplanted with leukemia cells from patients before and during sorafenib resistance recapitulated the clinical results. Both ITD and tyrosine kinase domain mutations at D835 were identified in leukemia initiating cells (LICs) from samples before sorafenib treatment. LICs bearing the D835 mutant have expanded during sorafenib treatment and dominated during the subsequent clinical resistance. These results suggest that sorafenib have selected more aggressive sorafenib-resistant subclones carrying both FLT3-ITD and D835 mutations, and might provide important leads to further improvement of treatment outcome with FLT3 inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzenesulfonates/administration & dosage , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Pyridines/administration & dosage , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Adult , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/adverse effects , Benzenesulfonates/adverse effects , Bone Marrow/enzymology , Bone Marrow/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Janus Kinase 3/biosynthesis , Janus Kinase 3/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Matrix Metalloproteinase 15/biosynthesis , Matrix Metalloproteinase 15/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Structure, Tertiary , Pyridines/adverse effects , Retinal Dehydrogenase , Sorafenib , Time Factors , Transplantation, Heterologous , fms-Like Tyrosine Kinase 3/metabolism
19.
Blood ; 119(25): 6089-98, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22446485

ABSTRACT

Interactions between the multikinase inhibitor sorafenib and the BH3-mimetic obatoclax (GX15-070) were examined in human acute myeloid leukemia (AML) cells. Treatment with sorafenib/obatoclax induced pronounced apoptosis in and reduced the clonogenic growth of multiple AML lines and primary AML cells but not normal CD34(+) cells. Sorafenib triggered rapid and pronounced Mcl-1 down-regulation accompanied by enhanced binding of Bim to Bcl-2 and Bcl-xL, effects that were abolished by obatoclax coadministration. Notably, shRNA knockdown of Bim, Bak, or Bax, but not Noxa, significantly attenuated obatoclax/sorafenib lethality, whereas ectopic expression of Mcl-1 exerted a protective effect. Furthermore, exposure of leukemia cells to sorafenib and obatoclax markedly induced autophagy, reflected by rapid and pronounced LC3 processing and LC3-green fluorescent protein (GFP) punctate formation. Multiple autophagy inhibitors or VPS34 knockdown, significantly potentiated sorafenib/obatoclax lethality, indicating a cytoprotective role for autophagy in this setting. Finally, studies in a xenograft mouse model revealed that combined sorafenib/obatoclax treatment markedly reduced tumor growth and significantly prolonged survival in association with Mcl-1 down-regulation and apoptosis induction, whereas agents administered individually had only modest effects. These findings suggest that combining sorafenib with agents that inhibit Mcl-1 and Bcl-2/Bcl-xL such as obatoclax may represent a novel and potentially effective strategy in AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/physiology , Apoptosis/drug effects , Benzenesulfonates/administration & dosage , Leukemia, Myeloid/drug therapy , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Pyridines/administration & dosage , Pyrroles/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Benzenesulfonates/pharmacology , Cells, Cultured , Drug Synergism , Female , HL-60 Cells , Humans , Indoles , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Membrane Proteins/metabolism , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyridines/pharmacology , Pyrroles/administration & dosage , Sorafenib , U937 Cells , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
20.
Dev Growth Differ ; 56(8): 555-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25280231

ABSTRACT

Fibroblast growth factor (FGF) signalling appears essential for the regulation of limb development, but a full complexity of this regulation remains unclear. Here, we addressed the effect of three different chemical inhibitors of FGF receptor tyrosine kinases (FGFR) on growth and patterning of the chicken wings. The inhibitor PD173074 caused shorter and thinner wing when using lower concentration. Microinjection of higher PD173074 concentrations (25 and 50 mmol/L) into the wing bud at stage 20 resulted in the development of small wing rudiment or the total absence of the wing. Skeletal analysis revealed the absence of the radius but not ulna, deformation of metacarpal bones and/or a reduction of digits. Treatment with PD161570 resembled the effects of PD173074. NF449 induced shortening and deformation of the developing wing with reduced autopodium. These malformed embryos mostly died at the stage HH25-29. PD173074 reduced chondrogenesis also in the limb micromass cultures together with early inhibition of cartilaginous nodule formation, evidenced by lack of sulphated proteoglycan and peanut agglutinin expression. The effect of FGFR inhibition on limb development observed here was unlikely mediated by excessive cell death as none of the inhibitors caused massive apoptosis at low concentrations. More probably, FGFR inhibition decreased both the proliferation and adhesion of mesenchymal chondroprogenitors. We conclude that FGFR signalling contributes to the regulation of the anterior-posterior patterning of zeugopod during chicken limb development.


Subject(s)
Benzenesulfonates/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Wings, Animal/drug effects , Wings, Animal/embryology , Animals , Benzenesulfonates/administration & dosage , Chick Embryo , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL