Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Control Release ; 353: 254-269, 2023 01.
Article in English | MEDLINE | ID: mdl-36442615

ABSTRACT

Over-activation of the endocannabinoid/CB1R system is a hallmark feature of obesity and its related comorbidities, most notably type 2 diabetes (T2D), and non-alcoholic fatty liver disease (NAFLD). Although the use of drugs that widely block the CB1R was found to be highly effective in treating all metabolic abnormalities associated with obesity, they are no longer considered a valid therapeutic option due to their adverse neuropsychiatric side effects. Here, we describe a novel nanotechnology-based drug delivery system for repurposing the abandoned first-in-class global CB1R antagonist, rimonabant, by encapsulating it in polymeric nanoparticles (NPs) for effective hepatic targeting of CB1Rs, enabling effective treatment of NAFLD and T2D. Rimonabant-encapsulated NPs (Rimo-NPs) were mainly distributed in the liver, spleen, and kidney, and only negligible marginal levels of rimonabant were found in the brain of mice treated by iv/ip administration. In contrast to freely administered rimonabant treatment, no CNS-mediated behavioral activities were detected in animals treated with Rimo-NPs. Chronic treatment of diet-induced obese mice with Rimo-NPs resulted in reduced hepatic steatosis and liver injury as well as enhanced insulin sensitivity, which were associated with enhanced cellular uptake of the formulation into hepatocytes. Collectively, we successfully developed a method of encapsulating the centrally acting CB1R blocker in NPs with desired physicochemical properties. This novel drug delivery system allows hepatic targeting of rimonabant to restore the metabolic advantages of blocking CB1R in peripheral tissues, especially in the liver, without the negative CB1R-mediated neuropsychiatric side effects.


Subject(s)
Cannabinoids , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Animals , Rimonabant/therapeutic use , Cannabinoid Receptor Antagonists/therapeutic use , Cannabinoid Receptor Antagonists/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Obesity/drug therapy , Cannabinoids/therapeutic use
2.
Pharmaceutics ; 13(12)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34959462

ABSTRACT

Multidrug resistance (MDR) of cancer cells remains a major obstacle to favorable outcomes of treatment with many drugs, including doxorubicin. Most of the clinical trials failed to demonstrate the benefit of the drug efflux transporter P-glycoprotein (P-gp) inhibitors to circumvent P-gp-mediated drug resistance in vivo. The present study explored the therapeutic potential of combined treatment with liposomal doxorubicin, P-gp inhibitor quinine, and the photodynamic therapy (PDT) using indocyanine green (ICG) in the adenocarcinoma drug-resistant tumor model. Liposomes were actively co-remotely loaded with doxorubicin and quinine, and ICG was passively adsorbed. The liposomes were characterized by differential scanning calorimetry (DSC) and cryogenic transmission microscopy (Cryo-TEM). We found that quinine impaired the crystalline structure of doxorubicin. In vitro, treatment with single agents themselves was insufficient to inhibit the growth of HT-29 MDR1 cells. However, pegylated liposomal doxorubicin and quinine (PLDQ) significantly diminished HT-29 MDR1 cell survival. Furthermore, survival inhibition intensified by the addition of ICG to the PLDQ (ICG + PLDQ). In vivo, ICG + PLDQ significantly decreased tumor growth when combined with tumor irradiation with NIR light (** p < 0.01). ICG + PLDQ + irradiation was superior to single treatments or combinational treatments without irradiation. These findings suggest that ICG + PLDQ can overcome P-gp-mediated MDR in cancer cells.

3.
Curr Drug Deliv ; 16(7): 637-644, 2019.
Article in English | MEDLINE | ID: mdl-31237208

ABSTRACT

BACKGROUND: Inflammation is a hallmark of epileptogenic brain tissue. Previously, we have shown that inflammation in epilepsy can be delineated using systemically-injected fluorescent and magnetite- laden nanoparticles. Suggested mechanisms included distribution of free nanoparticles across a compromised blood-brain barrier or their transfer by monocytes that infiltrate the epileptic brain. OBJECTIVE: In the current study, we evaluated monocytes as vehicles that deliver nanoparticles into the epileptic brain. We also assessed the effect of epilepsy on the systemic distribution of nanoparticleloaded monocytes. METHODS: The in vitro uptake of 300-nm nanoparticles labeled with magnetite and BODIPY (for optical imaging) was evaluated using rat monocytes and fluorescence detection. For in vivo studies we used the rat lithium-pilocarpine model of temporal lobe epilepsy. In vivo nanoparticle distribution was evaluated using immunohistochemistry. RESULTS: 89% of nanoparticle loading into rat monocytes was accomplished within 8 hours, enabling overnight nanoparticle loading ex vivo. The dose-normalized distribution of nanoparticle-loaded monocytes into the hippocampal CA1 and dentate gyrus of rats with spontaneous seizures was 176-fold and 380-fold higher compared to the free nanoparticles (p<0.05). Seizures were associated with greater nanoparticle accumulation within the liver and the spleen (p<0.05). CONCLUSION: Nanoparticle-loaded monocytes are attracted to epileptogenic brain tissue and may be used for labeling or targeting it, while significantly reducing the systemic dose of potentially toxic compounds. The effect of seizures on monocyte biodistribution should be further explored to better understand the systemic effects of epilepsy.


Subject(s)
Drug Delivery Systems , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Magnetite Nanoparticles/administration & dosage , Monocytes , Animals , Boron Compounds/administration & dosage , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Fluorescent Dyes/administration & dosage , Inflammation/metabolism , Kidney/metabolism , Lithium Chloride , Liver/metabolism , Male , Pilocarpine , Rats, Wistar , Spleen/metabolism
4.
Epilepsy Res ; 142: 64-72, 2018 05.
Article in English | MEDLINE | ID: mdl-29571151

ABSTRACT

Folate is involved in metabolic processes and it has been implicated in both aggravation and amelioration of seizures. The aim of the current work was to study the effect of chronic temporal lobe epilepsy (TLE) on the plasma and brain concentrations of folate and on its uptake carriers in the brain - the reduced folate carrier (RFC), folate receptor α (FRα) and proton coupled folate transporter (PCFT). We utilized the rat lithium pilocarpine model for TLE. Approximately two months following status epilepticus, rats with spontaneous recurrent seizures (SRS) were sacrificed for brain and plasma folate concentration analyses and folate uptake carrier expression studies. RT-PCR and western blot analyses were utilized for quantification of folate carriers' mRNAs and proteins, respectively. The distribution of folate carriers in the brain was studied using immunohistochemistry. In the SRS rats we found lower plasma concentrations (10 ±â€¯0.9 in control vs. 6.6 ±â€¯1.6 ng/ml in SRS, P < 0.05), but preserved cortical and increased hippocampal levels of folate (0.5 ±â€¯0.1 in control vs. 0.9 ±â€¯0.2 ng/mg in SRS, P = 0.055). Hippocampus - to - plasma ratio of folate concentration was 3-fold higher in the SRS group, compared with the controls (0.13 ±â€¯0.03 vs. 0.04 ±â€¯0.02, respectively; P < 0.01). mRNA and protein levels of the folate uptake carriers did not differ between SRS rats and controls. However, immunofluorescent staining quantification revealed that the emission intensity of both RFC and FRα was elevated 8-fold and 4-fold, respectively, in hippocampal CA1 neurons of SRS rats, compared to controls (P < 0.01). PCFT was unquantifiable. If corroborated by complementary research in humans, the findings of this study may be utilized clinically for supplemental therapy planning, in imaging the epileptic focus, and for drug delivery into the epileptic brain. Further studies are required for better elucidating the clinical and mechanistic significance of altered folate balances in the epileptic brain.


Subject(s)
Brain/metabolism , Folic Acid/metabolism , Homeostasis/physiology , Status Epilepticus/metabolism , Animals , CD11b Antigen/metabolism , Convulsants/toxicity , Disease Models, Animal , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folic Acid/genetics , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Homeostasis/drug effects , Lithium/toxicity , Male , Phosphopyruvate Hydratase/metabolism , Pilocarpine/toxicity , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Statistics, Nonparametric , Status Epilepticus/chemically induced , Status Epilepticus/pathology
5.
J Control Release ; 257: 2-9, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28215670

ABSTRACT

Antitumor therapy in the elderly is particularly challenging due to multiple, often chronic diseases, poly-therapy, and age-related physiological changes that affect drug efficacy and safety. Furthermore, tumors may become more aggressive and drug-resistant with advanced age, leading to poor patient prognosis. In this study, we evaluated in mice bearing medulloblastoma xenografts the effect of age on tumor progression and tumor therapy. We focused on therapeutic efficacy of two treatment modalities alone radiofrequency ablation therapy (RFA), PEGylated liposomal doxorubicin (PLD) equivalent to Doxil, and their combination. We demonstrated that tumor growth rate was higher and survival was lower in old versus young mice (p<0.05). Likewise, tumors in old mice were less susceptible to either PLD or RFA monotherapy. However, combined therapy of PLD and RFA succeeded to eliminate the age-related differences in anti-cancer treatment efficacy (p>0.05) by the two monotherapies. The results on PLD therapy are supported by preferable PEGylated nano-liposomes accumulation in tumors of young mice compared to old mice, as determined by near-infrared imaging with indocyanine green (ICG)-labeled PEGylated nano-liposomes. Taken together, our findings suggest that age effects on tumor progression and tumor monotherapy outcome may potentially be related to changes in tumor microenvironment, and that these changes can be overcome by RFA as this technique abolishes these differences and significantly improves success of PLD treatment.


Subject(s)
Aging , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/analogs & derivatives , Neoplasms/drug therapy , Neoplasms/surgery , Animals , Catheter Ablation , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Doxorubicin/therapeutic use , Humans , Mice , Mice, Nude , Models, Molecular , Neoplasms/pathology , Polyethylene Glycols/therapeutic use , Xenograft Model Antitumor Assays
6.
Front Pharmacol ; 7: 426, 2016.
Article in English | MEDLINE | ID: mdl-27895581

ABSTRACT

Aim: The multidrug resistance protein 1 (MDR1; P-glycoprotein) has been associated with efflux of chemotherapeutic agents from tumor cells and with poor patient prognosis. This study evaluated the feasibility of non-invasive, non-radioactive near infrared (NIR) imaging methodology for detection of MDR1 functional activity in tumors. Methods: Initial accumulation assays were conducted in MDR1-overexpressing MDCK cells (MDCK-MDR1) and control MDCK cells (MDCK-CT) using the NIR dyes indocyanine green (ICG), IR-783, IR-775, rhodamine 800, XenoLight DiR, and Genhance 750, at 0.4 µM-100 µM. ICG and IR-783 were also evaluated in HT-29 cells in which MDR1 overexpression was induced by colchicine (HT-29-MDR1) and their controls (HT-29-CT). In vivo optical imaging studies were conducted using immunodeficient mice bearing HT-29-CT and HT-29-MDR1 xenografts. Results: ICG's emission intensity was 2.0- and 2.2-fold higher in control versus MDR1-overexpressing cells, in MDCK and HT-29 cell lines, respectively. The respective IR-783 control:MDR1 ratio was 1.4 in both MDCK and HT-29 cells. Optical imaging of mice bearing HT-29-CT and HT-29-MDR1 xenografts revealed a statistically non-significant, 1.7-fold difference (p > 0.05) in ICG emission intensity between control and MDR1 tumors. No such differences were observed with IR-783. Conclusion: ICG and IR-783 appear to be weak MDR1 substrates. In vivo, low sensitivity and high between-subject variability impair the ability to use the currently studied probes as markers of tumor MDR1 activity. The results suggest that, for future use of this technology, additional NIR probes should be screened as MDR1 substrates.

7.
Epilepsia ; 57(6): e108-12, 2016 06.
Article in English | MEDLINE | ID: mdl-27142887

ABSTRACT

Our aim was to evaluate the effects of valproic acid (VPA) on the function of the placental barrier in vivo, in pregnant mice. Studies were conducted on gestational days 12.5 (mid-gestation) or 17.5 (late gestation), following intraperitoneal treatment with 200 mg/kg VPA or the vehicle. Indocyanine green (ICG; 0.167 mg, i.v.) was used as a marker for the placental barrier permeability. Transporter expression was evaluated by quantitative -PCR. VPA treatment was associated with a 40% increase (p < 0.05) in accumulation of ICG in maternal liver in mid-pregnancy and a decrease by one fifth (p < 0.05) in late pregnancy. Ex vivo, VPA treatment led to a 20% increase (p < 0.05) in fetal ICG emission in mid-pregnancy. Also in mid-pregnancy, the placental expression of the L-type amino acid transporter, the organic anion-transporting polypeptide (Oatp)4a1 (thyroid hormone transporter), and the reduced folate carrier was lower in VPA-treated mice (p < 0.05). In late pregnancy, hepatic Oatp4a1 levels were 40% less than in controls (p > 0.05). The observed changes in placental transporter expression and function support further research into the potential role of the placenta in the adverse pregnancy outcomes of VPA. Near-infrared imaging provides a noninvasive, nonradioactive tool for future studies on the effects of epilepsy and antiepileptic drugs on tissue transport functions.


Subject(s)
Anticonvulsants/pharmacology , Gene Expression Regulation/drug effects , Membrane Transport Proteins/metabolism , Placenta/drug effects , Valproic Acid/pharmacology , Age Factors , Animals , Female , Fetus/drug effects , Gestational Age , Indocyanine Green , Mice , Optical Imaging , Placenta/physiopathology , Pregnancy
8.
Nanomedicine ; 12(5): 1335-45, 2016 07.
Article in English | MEDLINE | ID: mdl-26964483

ABSTRACT

Correct localization of epileptic foci can improve surgical outcome in patients with drug-resistant seizures. Our aim was to demonstrate that systemically injected nanoparticles identify activated immune cells, which have been reported to accumulate in epileptogenic brain tissue. Fluorescent and magnetite-labeled nanoparticles were injected intravenously to rats with lithium-pilocarpine-induced chronic epilepsy. Cerebral uptake was studied ex vivo by confocal microscopy and MRI. Cellular uptake and biological effects were characterized in vitro in murine monocytes and microglia cell lines. Microscopy confirmed that the nanoparticles selectively accumulate within myeloid cells in the hippocampus, in association with inflammation. The nanoparticle signal was also detectable by MRI. The in vitro studies demonstrate rapid nanoparticle uptake and good cellular tolerability. We show that nanoparticles can target myeloid cells in epileptogenic brain tissue. This system can contribute to pre-surgical and intra-surgical localization of epileptic foci, and assist in detecting immune system involvement in epilepsy.


Subject(s)
Brain , Epilepsy/surgery , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Animals , Hippocampus , Humans , Inflammation , Mice , Microscopy, Confocal , Rats
9.
Theranostics ; 6(2): 167-76, 2016.
Article in English | MEDLINE | ID: mdl-26877776

ABSTRACT

Cerebral malaria (CM) is a major cause of death of Plasmodium falciparum infection. Misdiagnosis of CM often leads to treatment delay and mortality. Conventional brain imaging technologies are rarely applicable in endemic areas. Here we address the unmet need for a simple, non-invasive imaging methodology for early diagnosis of CM. This study presents the diagnostic and therapeutic monitoring using liposomes containing the FDA-approved fluorescent dye indocyanine green (ICG) in a CM murine model. Increased emission intensity of liposomal ICG was demonstrated in comparison with free ICG. The Liposomal ICG's emission was greater in the brains of the infected mice compared to naïve mice and drug treated mice (where CM was prevented). Histological analyses suggest that the accumulation of liposomal ICG in the cerebral vasculature is due to extensive uptake mediated by activated phagocytes. Overall, liposomal ICG offers a valuable diagnostic tool and a biomarker for effectiveness of CM treatment, as well as other diseases that involve inflammation and blood vessel occlusion.


Subject(s)
Fluorescent Dyes/pharmacokinetics , Indocyanine Green/pharmacokinetics , Liposomes/pharmacokinetics , Malaria, Cerebral/diagnosis , Animals , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Cell Line , Fluorescent Dyes/chemistry , Indocyanine Green/chemistry , Liposomes/chemistry , Malaria, Cerebral/drug therapy , Male , Mice , Mice, Inbred C57BL , Neuroimaging/methods
10.
Mol Pharm ; 12(9): 3351-7, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26149724

ABSTRACT

The transfer of indocyanine green (ICG) across the placenta is considered to be very low based on measurements in fetal blood. The goal of this study was to evaluate in mice ICG's distribution within fetuses themselves and effects of concomitant medications on fetal exposure. Mid-gestational (day 12.5) and late-gestational (day 17.5) age mice were imaged after administration of ICG (0.167 mg), in the presence and the absence of the organic anion transporting polypeptide (OATP) inhibitor rifampin (10 mg/kg, n = 11, or 20 mg/kg, n = 1) or the P-glycoprotein inhibitor valspodar (12.5 mg/kg). In vivo ICG emission intensity was followed by ex vivo analysis of blood and tissue emission. Both valspodar and rifampin increased ICG's emission intensity within maternal tissues. In addition, valspodar enhanced the ex vivo signal in mid-pregnancy placentae (2.1-fold; p < 0.01) and fetuses (2.4-fold; p < 0.01), and reduced late-pregnancy placenta:blood and fetus:blood ratios. Rifampin increased placental (1.4-fold, p < 0.05, and 2.3-fold, p < 0.01, in mid- and late-pregnancy, respectively) and fetal (2.2-fold, p < 0.01, and 3.2-fold, p < 0.01, in mid- and late-pregnancy) ICG signal. Similarly to valspodar, late-pregnancy placenta:blood and fetus:blood ratios were reduced by rifampin. Both inhibitors enhanced ICG's emission in fetal leg, liver, and brain. In conclusion, ICG distribution into the mouse fetus can be enhanced when used concomitantly with OATP or P-glycoprotein inhibitors. The greater distribution within individual fetal tissues is likely related to ICG's greater transplacental transfer. Until further data are available on ICG's safety when combined with medications that affect its maternal handling, such combinations should be used with caution.


Subject(s)
Cyclosporins/pharmacology , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Placenta/cytology , Placenta/metabolism , Rifampin/pharmacology , Spectroscopy, Near-Infrared/methods , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Antibiotics, Antitubercular/pharmacology , Coloring Agents/chemistry , Coloring Agents/pharmacokinetics , Diagnostic Imaging , Female , Mice , Organic Anion Transporters/antagonists & inhibitors , Placenta/drug effects , Pregnancy , Tissue Distribution
11.
Nanomedicine ; 11(5): 1057-64, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25791809

ABSTRACT

Intraoperative ureter identification can assist in the prevention of ureteral injury and consequently improve surgery outcomes. Our aim was to take advantage of the altered pharmacokinetics of liposomal indocyanine green (ICG), the only FDA-approved near-infrared (NIR) dye, for imaging of ureters during surgeries. ICG was passively adsorbed to liposomes. NIR whole mice body and isolated tissue imaging were used to study liposomal ICG properties vs. free ICG. In vivo, the urinary bladder could be clearly observed in most of the liposome-treated mice. Liposomal encapsulation of ICG enhanced ureteral emission up to 1.9 fold compared to free ICG (P<0.01). Increase in liposomal micropolarity and microviscosity and differential scanning calorimetry supported ICG localization within the liposomal bilayer. Our findings suggest that liposomal ICG could be utilized for ureteral imaging intra-operatively, thus potentially improving surgical outcomes. FROM THE CLINICAL EDITOR: Iatrogenic ureteral injury is a serious complication of abdominal surgery and intra-operative recognition of the ureters is usually the best method of injury prevention. In this article, the authors developed liposomal indocyanine green, which could be excreted via the urinary system and investigated its in-vivo use in mice.


Subject(s)
Coloring Agents/administration & dosage , Indocyanine Green/administration & dosage , Optical Imaging/methods , Ureter/pathology , Urinary Bladder/pathology , Animals , Coloring Agents/pharmacokinetics , Female , Indocyanine Green/pharmacokinetics , Infrared Rays , Liposomes , Mice , Ureter/injuries , Urinary Bladder/injuries
12.
Mol Pharm ; 9(12): 3595-601, 2012 Dec 03.
Article in English | MEDLINE | ID: mdl-23098218

ABSTRACT

The efflux transporter P-glycoprotein (P-gp) affects the pharmacokinetics of many drugs. Currently used methods for characterization of P-gp's functional activity in vivo involve the use of radiolabeled substrates, are costly, and are technically demanding. Our objective was to evaluate whether the FDA-approved near-infrared compound indocyanine green (ICG) can be used as a probe substrate of P-gp. We also characterized the interaction of ICG with another efflux transporter, the breast cancer resistance protein (BCRP). We evaluated ICG accumulation and transport in MDCK cells overexpressing P-gp or BCRP (MDCK-MDR1 and MDCK-BCRP, respectively) compared to control MDCK cells, in the presence or the absence of transporter inhibitors. In vivo imaging of ICG biodistribution in mice was conducted over 3.5 h using valspodar as the P-gp inhibitor. The EC50 values for ICG accumulation in control MDCK and MDCK-MDR1 cells were 9.0 × 10(-6) ± 5.7 × 10(-7) M and 1.5 × 10(-5) ± 1.1 × 10(-6) M, respectively. The efflux ratio for ICG in MDCK-MDR1 cells was 6.8-fold greater than in control cells. P-gp inhibition attenuated ICG efflux from MDR1-MDCK cells, and their effects in those cells were greater than in control MDCK cells. In contrast, BCRP level of expression or pharmacological inhibition did not significantly affect ICG cellular accumulation. In vivo imaging indicated enhanced cerebral ICG distribution with valspodar (brain - foot area under the concentration-time curves of 3.0 × 10(10), 5.6 × 10(10) and 3.7 × 10(10) h·[p/s/sr]/µW in valspodar-treated mice vs 9.0 × 10(9) and 5.3 × 10(9) h·[p/s/sr]/µW in controls). The findings from this pilot study suggest that near-infrared imaging using ICG as the probe substrate should be further characterized as a methodology for in vivo evaluation of P-gp activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/metabolism , Coloring Agents/metabolism , Indocyanine Green/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Blotting, Western , Cyclosporins/pharmacology , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Pilot Projects , Spectroscopy, Near-Infrared , Tissue Distribution
13.
Nanomedicine ; 7(4): 480-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21272665

ABSTRACT

A new liposome-based near-infrared probe that combines both imaging and targeting abilities was developed for application in medical imaging. The near-infrared fluorescent molecule indocyanine green (ICG), and the cetuximab monoclonal antibody for epidermal growth factor receptor (EGFR) were attached to liposomes by passive adsorption. It was found that ICG molecules adsorbed to the liposomes are more fluorescent than free ICG and have a larger quantum yield. Cetuximab-adsorbed fluorescent liposomes preserved EGFR recognition, as is evident from internalization and selective binding to A431 colon carcinoma cells overexpressing EGFR. The binding of cetuximab-targeted fluorescent liposomes to A431 compared with IEC-6 cells (normal enterocytes expressing physiological EGFR levels) was greater by a factor of 3.5, ensuring imaging abilities with available fluorescent equipment. Due to relatively high quantum yield and specific tumor cell-recognizing ability, this technology deserves further in vivo evaluation for imaging and diagnostic purposes. FROM THE CLINICAL EDITOR: A new liposome-based near-infrared probe combining both imaging and targeting abilities is reported. Due to relatively high quantum yield and EGFR-expressing tumor cell specificity, this technology deserves further in vivo evaluation for imaging and diagnostic purposes.


Subject(s)
Antibodies, Monoclonal/chemistry , Diagnostic Imaging/methods , Spectroscopy, Near-Infrared/methods , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cryoelectron Microscopy , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/metabolism , Humans , Indocyanine Green/chemistry , Liposomes/chemistry , Microscopy, Confocal , Microscopy, Electron, Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...