Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioorg Chem ; 142: 106967, 2024 01.
Article in English | MEDLINE | ID: mdl-37979321

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) or progeria is a rare genetic disease that causes premature aging, leading to a drastic reduction in the life expectancy of patients. Progeria is mainly caused by the intracellular accumulation of a defective protein called progerin, generated from a mutation in the LMNA gene. Currently, there is only one approved drug for the treatment of progeria, which has limited efficacy. It is believed that progerin levels are the most important biomarker related to the severity of the disease. However, there is a lack of effective tools to directly visualize progerin in the native cellular models, since the commercially available antibodies are not well suited for the direct visualization of progerin in cells from the mouse model of the disease. In this context, an alternative option for the visualization of a protein relies on the use of fluorescent chemical probes, molecules with affinity and specificity towards a protein. In this work we report the synthesis and characterization of a new fluorescent probe (UCM-23079) that allows for the direct visualization of progerin in cells from the most widely used progeroid mouse model. Thus, UCM-23079 is a new tool compound that could help prioritize potential preclinical therapies towards the final goal of finding a definitive cure for progeria.


Subject(s)
Progeria , Mice , Animals , Humans , Progeria/drug therapy , Progeria/genetics , Progeria/metabolism , Fluorescent Dyes/therapeutic use , Mutation
2.
Eur J Nucl Med Mol Imaging ; 50(6): 1765-1779, 2023 05.
Article in English | MEDLINE | ID: mdl-36692541

ABSTRACT

PURPOSE: The once highly anticipated antibody-based pathway-targeted therapies have not achieved promising outcomes for deadly pancreatic ductal adenocarcinoma (PDAC), mainly due to drugs' low intrinsic anticancer activity and poor penetration across the dense physiological barrier. This study aims to develop an ultra-small-sized, EGFR/VEGF bispecific therapeutic protein to largely penetrate deep tumor tissue and effectively inhibit PDAC tumor growth in vivo. METHODS: The bispecific protein, Bi-fp50, was constructed by a typical synthetic biology method and labeled with fluorescent dyes for in vitro and in vivo imaging. Physicochemical properties, protein dual-binding affinity, and specificity of the Bi-fp50 were evaluated in several PDAC cell lines. In vitro quantitatively and qualitatively anticancer activity of Bi-fp50 was assessed by live/dead staining, MTT assay, and flow cytometry. In vivo pharmacokinetic and biodistribution were evaluated using blood biopsy samples and near-infrared fluorescence imaging. In vivo real-time tracking of Bi-fp50 in the local tumor was conducted by fibered confocal fluorescence microscopy. The subcutaneous PDAC tumor model was used to assess the in vivo antitumor effect of Bi-fp50. RESULTS: Bi-fp50 with an ultra-small size of 50 kDa (5 ~ 6 nm) showed an excellent binding ability to VEGF and EGFR simultaneously and had enhanced, accumulated binding capability for Bxpc3 PDAC cells compared with anti-VEGF scFv and anti-EGFR scFv alone. Additionally, bi-fp50 significantly inhibited the proliferation and growth of Bxpc3 and Aspc1 PDAC cells even under a relatively low concentration (0.3 µM). It showed synergistically enhanced therapeutic effects relative to two individual scFv and Bi-fp50x control in vitro. The half-life of blood clearance of Bi-fp50 was 4.33 ± 0.23 h. After intravenous injection, Bi-fp50 gradually penetrated the deep tumor, widely distributed throughout the whole tissue, and primarily enriched in the tumor with nearly twice the accumulation than scFv2 in the orthotopic PDAC tumor model. Furthermore, the Bi-fp50 protein could induce broad apoptosis in the whole tumor and significantly inhibited tumor growth 3 weeks after injection in vivo without other noticeable side effects. CONCLUSION: The proof-of-concept study demonstrated that the ultra-small-sized, bispecific protein Bi-fp50 could be a potential tumor suppressor and an efficient, safe theranostic tool for treating PDAC tumors.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tissue Distribution , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/therapy , Fluorescent Dyes/therapeutic use , Cell Line, Tumor , Pancreatic Neoplasms
3.
J Am Chem Soc ; 144(49): 22562-22573, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36445324

ABSTRACT

Restoring innate apoptosis and simultaneously inhibiting metastasis by a molecular drug is an effective cancer therapeutic approach. Herein, a large rigid and V-shaped NIR-II dye, DUT850, is rationally designed for potential cardiolipin (CL)-targeted chemo-phototheranostic application. DUT850 displays moderate NIR-II fluorescence, excellent photodynamic therapy (PDT) and photothermal therapy (PTT) performance, and ultra-high photostability. More importantly, the unique rigid V-shaped backbone, positive charge, and lipophilicity of DUT850 afford its specific recognition and efficient binding to CL; such an interaction of DUT850-CL induced a spectrum of physiological disruptions, including translocation of cytochrome c, Ca2+ overload, reactive oxygen species burst, and ATP depletion, which not only activated cancer cell apoptosis but also inhibited tumor metastasis both in vitro and in vivo. Furthermore, the tight binding of DUT850-CL improves the phototoxicity of DUT850 toward cancer cells (IC50 as low as 90 nM) under safe 808 nm laser irradiation (330 mW cm-2). Upon encapsulation into bovine serum albumin (BSA), DUT850@BSA exerted a synergetic chemo-PDT-PTT effect on the 4T1 tumor mouse model, eventually leading to solid tumor annihilation and metastasis inhibition, which could be followed in real time with the NIR-II fluorescence of DUT850. This work contributed a promising approach for simultaneously re-engaging cancer cell apoptotic networks and activating the anti-metastasis pathway by targeting a pivotal upstream effector, which will bring a medical boon for inhibition of tumor proliferation and metastasis.


Subject(s)
Avalanches , Nanoparticles , Neoplasms , Photochemotherapy , Mice , Animals , Phototherapy , Cardiolipins , Neoplasms/drug therapy , Fluorescent Dyes/therapeutic use , Serum Albumin, Bovine/chemistry , Apoptosis , Nanoparticles/chemistry , Cell Line, Tumor
4.
Anal Chem ; 94(6): 2820-2826, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35119275

ABSTRACT

In this paper, we propose a hydrogel-coated gate field-effect transistor (FET) for the real-time and label-free monitoring of ß-amyloid (Aß) aggregation and its inhibition. The hydrogel used in this study is composed of poly tetramethoxysilane (TMOS), in which Aß monomers are entrapped and then aggregate, and coated on the gate insulator; that is, Aß aggregation is induced in the vicinity of the sensing surface. With the Aß hydrogel-coated gate FET, the steplike decrease in the surface potential of the Aß hydrogel-coated gate electrode is electrically monitored in real time, according to the stepwise aggregation of Aß monomers to form into fibrils through oligomers and so forth in stages. This is because the capacitance of the Aß-hydrogel membrane decreases depending on the stage of aggregation; that is, the hydrophobicity of the Aß-hydrogel membrane increases stepwise depending on the amount of Aß aggregates. The formation of Aß fibrils is also confirmed in the measurement solution using a fluorescent dye, thioflavin T, which selectively binds to the Aß fibrils. Moreover, the addition of daunomycin, an inhibitor of Aß aggregation, to the measurement solution suppresses the stepwise electrical response of the Aß hydrogel-coated gate FET. Thus, a platform based on the Aß hydrogel-coated gate FET is suitable for a simple screening system for inhibitors of Aß aggregation, which may lead the identification of potential therapeutic agents for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Fluorescent Dyes/therapeutic use , Humans , Hydrogels , Hydrophobic and Hydrophilic Interactions , Peptide Fragments/metabolism
5.
Anal Chem ; 94(9): 4072-4077, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35194985

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fatal interstitial pneumonia with unknown pathogenesis. Early diagnosis and therapeutic intervention are essential for improving the prognosis of patients with IPF. The level of nitric oxide upregulates in the alveoli of IPF patients, which is correlated with the severity of the disease. Herein, we report a fluorescent probe DCM-nitric oxide (NO) to detect IPF by monitoring the concentration changes of NO. This probe displays a fast response time and a good linear response to NO in vitro. Fluorescence imaging experiments with probe DCM-NO revealed that the level of intracellular NO increases in the pulmonary fibrosis cells and IPF mice models. Probe DCM-NO displayed a strong red fluorescence in IPF mice models. However, a declining fluorescence was evidenced in the OFEV-treated IPF mice, implying that DCM-NO is capable of evaluating the therapeutic effects on IPF. Thus, probe DCM-NO can quickly predict the progression of pulmonary fibrosis at an early stage and thus help improve the effective treatment.


Subject(s)
Idiopathic Pulmonary Fibrosis , Animals , Fluorescent Dyes/therapeutic use , Humans , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Idiopathic Pulmonary Fibrosis/drug therapy , Mice , Nitric Oxide , Optical Imaging , Pulmonary Alveoli/pathology
6.
Anal Chem ; 94(39): 13498-13506, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36121878

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disease that makes the brain nervous system degenerate rapidly and is accompanied by some special cognitive and behavioral dysfunction. Recently, butyrylcholinesterase (BChE) was reported as an important enzyme, whose activity can provide predictive value for timely discovery and diagnosis of AD. Therefore, it is indispensable to design a detection tool for selective and rapid response toward BChE. In this study, we developed a novel near-infrared fluorescent probe (Chy-1) for the detection of BChE activity. An excellent sensitivity, good biocompatibility, and lower limit of detection (LOD) of 0.12 ng/mL made the probe extremely specific for BChE, which was successfully used in biological imaging. What is more, Chy-1 can not only clearly distinguish tumor from normal cells but also forms a clear boundary between the normal and cancer tissues due to the obvious difference in fluorescence intensity produced via in situ spraying. Most important of all, Chy-1 was also successfully applied to track the BChE activity in AD mouse models. Based on this research, the novel probe may be a powerful tool for clinical diagnosis and therapy of tumor and neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Alzheimer Disease/diagnostic imaging , Animals , Brain/metabolism , Butyrylcholinesterase/metabolism , Fluorescent Dyes/therapeutic use , Mice
7.
Clin Exp Allergy ; 52(9): 1059-1070, 2022 09.
Article in English | MEDLINE | ID: mdl-35278245

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) is a rare genetic disease that leads to recurrent episodes of swelling and pain caused by uncontrolled plasma kallikrein (PKa) activity. Current guidelines recommend ready availability of on-demand HAE treatments that can be administered early upon attack onset. This report describes the pharmacological and pharmacodynamic properties of the novel oral small-molecule PKa inhibitor KVD900 as a potential on-demand treatment for HAE. METHODS: Pharmacological properties of KVD900 on PKa and closely related serine proteases were characterized using kinetic fluorogenic substrate activity assays. Effects of KVD900 on PKa activity and kallikrein kinin system activation in whole plasma were measured in the presence of dextran sulphate (DXS)-stimulation using a fluorogenic substrate and capillary immunoassays to quantify high molecular weight kininogen (HK), plasma prekallikrein and Factor XII cleavage. Pharmacodynamic effects of orally administered KVD900 were characterized in plasma samples from six healthy controls in a first in human phase 1 clinical trial and from 12 participants with HAE in a phase 2 clinical trial. RESULTS: KVD900 is a selective, competitive and reversible inhibitor of human PKa enzyme with a Ki of 3.02 nM. The association constant (Kon ) of KVD900 for PKa is >10 × 106  M-1  s-1 . Oral administration of KVD900 in a first-in-human clinical trial achieved rapid and near complete inhibition of DXS-stimulated PKa enzyme activity and HK cleavage and reduced plasma prekallikrein and Factor XII activation in plasma. In individuals with HAE, orally administered KVD900 inhibited DXS-stimulated PKa activity in plasma by ≥95% from 45 min to at least 4 h post-dose and provided rapid protection of HK from cleavage. CONCLUSION: KVD900 is a fast-acting oral PKa inhibitor that rapidly inhibits PKa activity, kallikrein kinin system activation and HK cleavage in plasma. On-demand administration of KVD900 may provide an opportunity to halt the generation of bradykinin and reverse HAE attacks.


Subject(s)
Angioedemas, Hereditary , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/prevention & control , Bradykinin , Complement C1 Inhibitor Protein/genetics , Factor XII , Fluorescent Dyes/therapeutic use , Humans , Kallikrein-Kinin System , Plasma Kallikrein , Prekallikrein/metabolism
8.
Bioconjug Chem ; 33(5): 918-928, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35504859

ABSTRACT

The application of conventional fluorescent probes in living cells has been limited by excess fluorescence interference, reduced selectivity, and poor permeability. Herein, we describe a convenient solution for overcoming the above limitations based on bio-orthogonal reactions and releasable linkers that provide bifunctional molecules for imaging and therapeutic integration. To reduce the interference of excess fluorescent moieties, a bio-orthogonal reaction was applied to activate the fluorescence of the active parent drugs without fluorophores. Moreover, disulfide bonds were incorporated as releasable linkers. After imaging the target protein, the newly yielded fluorophore could be released from the active drugs based on the highly reducing conditions of the tumor. Thus, these bifunctional molecules are comparable in therapeutic activity to the parent drug. These novel imaging and therapeutic integration molecules could be used to realize imaging-aided diagnosis and perform efficient real-time monitoring of cancer cells. Our findings are expected to enable efficient and specific imaging and real-time in vivo prognostic monitoring in the clinic.


Subject(s)
Fluorescent Dyes , Neoplasms , Diagnostic Imaging , Disulfides/metabolism , Fluorescence , Fluorescent Dyes/chemistry , Fluorescent Dyes/therapeutic use , Humans , Neoplasms/diagnostic imaging , Neoplasms/drug therapy
9.
Anal Biochem ; 655: 114854, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35963341

ABSTRACT

The translocator protein 18 kDa (TSPO) was first identified in 1997, and has now become one of the appealing subcellular targets in medicinal chemistry and its related fields. TSPO involves in a variety of diseases, covering neurodegenerative diseases, psychiatric disorders, cancers, and so on. To date, various high-affinity TSPO ligands labelled with single-photon emission computed tomography (SPECT)/positron emission tomography (PET) radionuclides have been reported, with some third-generation radioligands advanced to clinical trials. On the other hand, only a few number of TSPO ligands have been labelled with fluorophores for disease diagnosis. It is noteworthy that the majority of the TSPO fluorescent probes synthesised to date are based on visible fluorophores, suggesting that their applications are limited to in vitro studies, such as in vitro imaging of cancer cells, post-mortem analysis, and tissue biopsies examinations. In this context, the potential application of TSPO ligands can be broadened for in vivo investigations of human diseases by labelling with near-infrared (NIR)-fluorophores or substituting visible fluorophores with NIR-fluorophores on the currently developed fluorescent probes. In this review article, recent progress on fluorescent probes targeting the TSPO are summarised, with an emphasis on development trend in recent years and application prospects in the future.


Subject(s)
Neurodegenerative Diseases , Receptors, GABA , Carrier Proteins , Fluorescent Dyes/therapeutic use , Humans , Ligands , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Positron-Emission Tomography/methods , Receptors, GABA/analysis , Receptors, GABA/metabolism , Receptors, GABA/therapeutic use
10.
Lasers Surg Med ; 54(6): 861-874, 2022 08.
Article in English | MEDLINE | ID: mdl-35451510

ABSTRACT

PURPOSE: Conventional oral antifungal therapies for onychomycosis (OM) often do not achieve complete cure and may be associated with adverse effects, medical interactions, and compliance issues restricting their use in a large group of patients. Topical treatment can bypass the systemic side effects but is limited by the physical barrier of the nail plate. Ablative fractional laser (AFL) treatment can be used to improve the penetration of topical drugs into the nail. This study visualized the effects of laser ablation of nail tissue and assessed their impact on the biodistribution of a fluorescent dye in healthy and fungal nail tissue. METHODS: For the qualitative assessment of CO2 AFL effects on healthy nail tissue, scanning electron microscopy (SEM), coherent anti-Stokes Raman scattering microscopy (CARS-M), and widefield fluorescence microscopy (WFM) were used. To quantitate the effect of laser-pretreatment on the delivery of a fluorescent dye, ATTO-647N, into healthy and fungal nail tissue, ablation depth, nail plate thickness, and ATTO-647N fluorescence intensity in three nail plate layers were measured using WFM. A total of 30 nail clippings (healthy n = 18, fungal n = 12) were collected. An aqueous ATTO-647N solution was directly applied to the dorsal surface of 24 nail samples (healthy n = 12, fungal n = 12) and incubated for 4 hours, of which half (healthy n = 6, fungal n = 6) had been pretreated with AFL (30 mJ/mb, 15% density, 300 Hz, pulse duration <1 ms). RESULTS: Imaging revealed a three-layered nail structure, an AFL-induced porous ablation crater, and changes in autofluorescence. While intact fungal samples showed a 106% higher ATTO-647N signal intensity than healthy controls, microporation led to a significantly increased fluorophore permeation in all samples (p < 0.0001). AFL processing of nail tissue enhanced topical delivery of ATTO-647N in all layers, (average increase: healthy +108%, fungal +33%), most pronounced in the top nail layer (healthy +122%, fungal +68%). While proportionally deeper ablation craters correlated moderately with higher fluorescence intensities in healthy nail tissue, fungal samples showed no significant relationship. CONCLUSION: Fractional CO2 laser microporation is a simple way of enhancing the passive delivery of topically applied ATTO-647N. Although the impaired nail plate barrier in OM leads to greater diffusion of the aqueous solution, AFL can increase the permeability of both structurally deficient and intact nails.


Subject(s)
Lasers, Gas , Onychomycosis , Administration, Topical , Carbon Dioxide/metabolism , Carbon Dioxide/pharmacology , Carbon Dioxide/therapeutic use , Fluorescent Dyes/therapeutic use , Humans , Lasers, Gas/therapeutic use , Nails , Onychomycosis/diagnostic imaging , Onychomycosis/surgery , Tissue Distribution
11.
Int J Mol Sci ; 23(21)2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36361667

ABSTRACT

A total of 20% to 50% of prostate cancer (PCa) patients leave the surgery room with positive tumour margins. The intraoperative combination of fluorescence guided surgery (FGS) and photodynamic therapy (PDT) may be very helpful for improving tumour margin delineation and cancer therapy. PSMA is a transmembrane protein overexpressed in 90−100% of PCa cells. The goal of this work is the development of a PSMA-targeted Near InfraRed Fluorescent probe to offer the surgeon a valuable intraoperative tool for allowing a complete tumour removal, implemented with the possibility of using PDT to kill the eventual not resected cancer cells. PSMA-617 binding motif was conjugated to IRDye700DX-NHS and the conjugation did not affect the photophysical characteristics of the fluorophore. The affinity of IRDye700DX-PSMA-617 towards PCa cells followed the order of their PSMA expression, i.e., PC3-PIP > LNCaP > PC3, PC3-FLU. NIRF imaging showed a significant PC3-PIP tumour uptake after the injection of 1 or 5 nmol with a maximum tumour-to-muscle ratio (ca. 60) observed for both doses 24 h post-injection. Importantly, urine, healthy prostate, and the bladder were not fluorescent at 24 h post-injection. Flow cytometry and confocal images highlighted a co-localization of PSMA+ cells with IRDye700DX-PSMA uptake. Very interestingly, ex vivo analysis on a tumour specimen highlighted a significant PSMA expression by tumour-associated macrophages, likely attributable to extracellular vesicles secreted by the PSMA(+) tumour cells. FGS proved that IRDye700DX-PSMA was able to easily delineate tumour margins. PDT experiments showed a concentration-dependent decrease in cell viability (from 75% at 10 nM to 12% at 500 nM), whereas controls did not show any cytotoxicity. PC3-PIP tumour-bearing mice subjected to photodynamic therapy showed a delayed tumour growth. In conclusion, a novel PSMA-targeted NIRF dye with dual imaging-PDT capabilities was synthesized and displayed superior specificity compared to other small PSMA targeted molecules.


Subject(s)
Photochemotherapy , Prostatic Neoplasms , Surgery, Computer-Assisted , Animals , Humans , Male , Mice , Antigens, Surface , Cell Line, Tumor , Fluorescent Dyes/pharmacology , Fluorescent Dyes/therapeutic use , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Surgery, Computer-Assisted/methods
12.
Molecules ; 27(18)2022 Sep 10.
Article in English | MEDLINE | ID: mdl-36144609

ABSTRACT

Upconversion (UC) is a process that describes the emission of shorter-wavelength light compared to that of the excitation source. Thus, UC is also referred to as anti-Stokes emission because the excitation wavelength is longer than the emission wavelength. UC materials are used in many fields, from electronics to medicine. The objective of using UC in medical research is to synthesize upconversion nanoparticles (UCNPs) composed of a lanthanide core with a coating of adsorbed dye that will generate fluorescence after excitation with near-infrared light to illuminate deep tissue. Emission occurs in the visible and UV range, and excitation mainly in the near-infrared spectrum. UC is observed for lanthanide ions due to the arrangement of their energy levels resulting from f-f electronic transitions. Organic compounds and transition metal ions are also able to form the UC process. Biocompatible UCNPs are designed to absorb infrared light and emit visible light in the UC process. Fluorescent dyes are adsorbed to UCNPs and employed in PDT to achieve deeper tissue effects upon irradiation with infrared light. Fluorescent UCNPs afford selectivity as they may be activated only by illumination of an area of diseased tissue, such as a tumor, with infrared light and are by themselves atoxic in the absence of infrared light. UCNP constructs can be monitored as to their location in the body and uptake by cancer cells, aiding in evaluation of exact doses required to treat the targeted cancer. In this paper, we review current research in UC studies and UCNP development.


Subject(s)
Lanthanoid Series Elements , Nanoparticles , Neoplasms , Fluorescent Dyes/therapeutic use , Infrared Rays , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photons
13.
J Am Chem Soc ; 143(49): 20828-20836, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34860505

ABSTRACT

In recent years, cancer phototherapy has been extensively studied as noninvasive cancer treatment. To present efficient recognition toward cancer cells, most photosensitizers (PSs) are required to couple with tumor-targeted ligands. Interestingly, the heptamethine cyanine IR780 displays an intrinsic tumor-targeted feature even without modification. However, the photothermal efficacy and photostability of IR780 are not sufficient enough for clinical use. Herein, we involve a twisted structure of tetraphenylethene (TPE) between two molecules of IR780 to improve the photothermal conversion efficiency (PCE). The obtained molecule T780T shows strong near-infrared (NIR) fluorescence and improved PCE (38.5%) in the dispersed state. Also, the photothermal stability and ROS generation capability of T780T at the NIR range (808 nm) are both promoted. In the aqueous phase, the T780T was formulated into uniform nanoaggregates (∼200 nm) with extremely low fluorescence and PTT response, which would reduce in vivo imaging background and side effect of PTT response in normal tissues. After intravenous injection into tumor-bearing mice, the T780T nanoaggregates display high tumor accumulation and thus remarkably inhibit the tumor growth. Moreover, the enhanced photostability of the T780T allows for twice irradiation after one injection and leads to more significant tumor inhibition. In summary, our study presents a tumor-targeted small-molecule PS for efficient cancer therapy and brings a new design of heptamethine cyanine PS for potential clinical applications.


Subject(s)
Antineoplastic Agents/therapeutic use , Fluorescent Dyes/therapeutic use , Indoles/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Stilbenes/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/radiation effects , Humans , Indoles/chemical synthesis , Indoles/radiation effects , Infrared Rays , Mice, Inbred BALB C , Neoplasms/diagnostic imaging , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Photothermal Therapy , Stilbenes/chemical synthesis , Stilbenes/radiation effects
14.
J Am Chem Soc ; 143(35): 14115-14124, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34374290

ABSTRACT

Breast cancer consists of heterogenic subpopulations, which determine the prognosis and response to chemotherapy. Among these subpopulations, a very limited number of cancer cells are particularly problematic. These cells, known as breast cancer stem cells (BCSCs), are thought responsible for metastasis and recurrence. They are thus major contributor to the unfavorable outcomes seen for many breast cancer patients. BCSCs are more prevalent in the hypoxic niche. This is an oxygen-deprived environment that is considered crucial to their proliferation, stemness, and self-renewal but also one that makes BCSCs highly refractory to traditional chemotherapeutic regimens. Here we report a small molecule construct, AzCDF, that allows the therapeutic targeting of BCSCs and which is effective in normally refractory hypoxic tumor environments. A related system, AzNap, has been developed that permits CSC imaging. Several design elements are incorporated into AzCDF, including the CAIX inhibitor acetazolamide (Az) to promote localization in MDA-MB-231 CSCs, a dimethylnitrothiophene subunit as a hypoxia trigger, and a 3,4-difluorobenzylidene curcumin (CDF) as a readily released therapeutic payload. This allows AzCDF to serve as a hypoxia-liable molecular platform that targets BCSCs selectively which decreases CSC migration, retards tumor growth, and lowers tumorigenesis rates as evidenced by a combination of in vitro and in vivo studies. To the best of our knowledge, this is the first time a CSC-targeting small molecule has been shown to prevent tumorigenesis in an animal model.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbonic Anhydrase Inhibitors/therapeutic use , Carcinogenesis/drug effects , Cell Hypoxia/drug effects , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Acetazolamide/analogs & derivatives , Acetazolamide/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Cell Line, Tumor , Cell Movement/drug effects , Curcumin/analogs & derivatives , Curcumin/chemical synthesis , Curcumin/therapeutic use , Diarylheptanoids/chemical synthesis , Diarylheptanoids/therapeutic use , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/therapeutic use , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasms/diagnostic imaging , Spheroids, Cellular/drug effects , Thiophenes/chemical synthesis , Thiophenes/therapeutic use , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
15.
Anal Biochem ; 612: 113900, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32926864

ABSTRACT

Extracellular pH plays vital roles in physiological and pathological processes including tumor metastasis and chemotherapy resistance. Abnormal extracellular pH is known to be associated with various pathological states, such as those in tumors, ischemic stroke, infection, and inflammation. Specifically, dysregulated pH is regarded as a hallmark of cancer because enhanced glycolysis and poor perfusion in most solid malignant tumors create an acidic extracellular environment, which enhances tumor growth, invasion, and metastasis. Close connection between the cell functions with extracellular pH means that precise and real-time measurement of the dynamic change of extracellular pH can provide critical information for not only studying physiological and pathological processes but also diagnosis of cancer and other diseases. This review highlights the recent development of based fluorescent probes for extracellular pH measurement, including design strategies, reaction mechanism and applications for the detection and imaging of extracellular pH.


Subject(s)
Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Optical Imaging/methods , Extracellular Space/diagnostic imaging , Fluorescent Dyes/therapeutic use , Humans , Hydrogen-Ion Concentration , Intracellular Space/diagnostic imaging , Neoplasms/chemistry , Neoplasms/diagnostic imaging , Neoplasms/pathology
16.
Future Oncol ; 17(9): 1083-1095, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33590768

ABSTRACT

Robot-assisted radical prostatectomy has become the standard of care for the removal of localized prostate cancer. Positive outcomes depend upon the precise removal of the prostate and associated tissue without damage to nearby structures. This process can be aided by fluorescence-guided surgery to enhance the visual contrast between different structures. Here the authors have conducted a systematic review using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify ten investigations into the use of fluorescence-guided surgery in robot-assisted radical prostatectomy. These studies used fluorescent tracers to identify structures, including the prostate, neurovascular bundle and lymph nodes. These studies demonstrate the safe and effective use of fluorescence-guided surgery in robot-assisted radical prostatectomy and pave the way for further developments in this field.


Subject(s)
Fluorescent Dyes/therapeutic use , Prostatectomy , Prostatic Neoplasms/surgery , Robotic Surgical Procedures , Fluorescence , Fluorescent Dyes/metabolism , Humans , Intraoperative Period , Lymph Nodes/metabolism , Lymph Nodes/surgery , Male , Organ Sparing Treatments , Prostate/innervation , Prostate/metabolism , Prostate/surgery
17.
Surg Endosc ; 35(5): 2373-2385, 2021 05.
Article in English | MEDLINE | ID: mdl-33495878

ABSTRACT

BACKGROUND: Intestinal perfusion at the anastomotic site is thought to be one of the most influential risk factors for postoperative anastomotic leakage (AL). We evaluated the efficacy of indocyanine green (ICG) fluorescence imaging at the stump of the proximal colon in left-sided colectomy or rectal resection in terms of decreasing the incidence of AL. METHODS: Prospectively collected data were retrospectively evaluated. Patients who underwent left-sided colectomy or rectal resection were enrolled (ICG group; n = 197), and patients who had undergone a similar procedure before the ICG group were enrolled from the charts as historical controls (HC group; n = 187). After ICG evaluation, anastomosis was performed where fluorescence was sufficient. The incidence of AL was compared between the ICG and HC groups. Propensity score (PS)-matched data were analyzed to clarify the risk of AL. RESULTS: AL occurred in 6 patients (3.3%) in the ICG group and 17 (10.7%) in the HC group. ICG evaluation revealed 179 patients with good fluorescence and 18 with poor/none perfusion (9.1%). The transection line was changed in all patients with poor/none fluorescence. Three of these 18 patients developed AL (16.7%), though transection line was changed at which is thought to be good. We hope AL in poor/none fluorescence can be prevented at the same rate of cases with good fluorescence. Actually, the rate of that was significantly higher compared with good fluorescence patients (P = 0.038). 93 patients in each group were compared by PS-matched data analysis, which showed the AL rate in the ICG group was significantly lower than that in the HC group (3.2% vs 10.8%, respectively; P = 0.046). CONCLUSIONS: Even though this study has limitations of comparison of data prospectively collected and retrospectively analyzed, intraoperative ICG fluorescence imaging evaluation could significantly decrease the incidence of AL.


Subject(s)
Anastomotic Leak/prevention & control , Colonic Neoplasms/surgery , Indocyanine Green/therapeutic use , Optical Imaging/methods , Rectal Neoplasms/surgery , Adult , Aged , Aged, 80 and over , Anastomosis, Surgical/adverse effects , Anastomosis, Surgical/methods , Anastomotic Leak/diagnostic imaging , Anastomotic Leak/etiology , Colectomy/adverse effects , Colectomy/methods , Colon/surgery , Female , Fluorescent Dyes/therapeutic use , Humans , Intraoperative Care/methods , Male , Middle Aged , Perfusion , Proctectomy/adverse effects , Propensity Score , Prospective Studies
18.
Surg Endosc ; 35(5): 2091-2103, 2021 05.
Article in English | MEDLINE | ID: mdl-32405892

ABSTRACT

BACKGROUND: Confocal laser endomicroscopy (CLE) is a novel endoscopic adjunct that allows real-time in vivo histological examination of mucosal surfaces. By using intravenous or topical fluorescent agents, CLE highlights certain mucosal elements that facilitate an optical biopsy in real time. CLE technology has been used in different organ systems including the gastrointestinal tract. There has been numerous studies evaluating this technology in gastrointestinal endoscopy, our aim was to evaluate the safety, value, and efficacy of this technology in the gastrointestinal tract. METHODS: The Society of American Gastrointestinal and Endoscopic Surgeons (SAGES) Technology and Value Assessment Committee (TAVAC) performed a PubMed/Medline database search of clinical studies involving CLE in May of 2018. The literature search used combinations of the keywords: confocal laser endomicroscopy, pCLE, Cellvizio, in vivo microscopy, optical histology, advanced endoscopic imaging, and optical diagnosis. Bibliographies of key references were searched for relevant studies not covered by the PubMed search. Case reports and small case series were excluded. The manufacturer's website was also used to identify key references. The United States Food and Drug Administration (U.S. FDA) Manufacturer And User facility and Device Experience (MAUDE) database was searched for reports regarding the device malfunction or injuries. RESULTS: The technology offers an excellent safety profile with rare adverse events related to the use of fluorescent agents. It has been shown to increase the detection of dysplastic Barrett's esophagus, gastric intraepithelial neoplasia/early gastric cancer, and dysplasia associated with inflammatory bowel disease when compared to standard screening protocols. It also aids in the differentiation and classification of colorectal polyps, indeterminate biliary strictures, and pancreatic cystic lesions. CONCLUSIONS: CLE has an excellent safety profile. CLE can increase the diagnostic accuracy in a number of gastrointestinal pathologies.


Subject(s)
Endoscopy, Gastrointestinal/instrumentation , Endoscopy, Gastrointestinal/methods , Microscopy, Confocal/methods , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/pathology , Early Detection of Cancer , Endoscopy, Gastrointestinal/adverse effects , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/therapeutic use , Humans , Lasers , Microscopy, Confocal/instrumentation , Pancreas/diagnostic imaging , Pancreas/pathology , Practice Guidelines as Topic , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology
19.
J Nanobiotechnology ; 19(1): 37, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33541369

ABSTRACT

BACKGROUND: The aim to develop a highly stable near-infrared (NIR) photoinduced tumor therapy agent stems from its considerable potential for biological application. Due to its long wavelength, biological imaging exhibits a high signal-to-background ratio, deep tissue penetration and maximum permissible light power, which can minimize damage to an organism during photoinduced tumor therapy. RESULTS: A class of stable NIR-II fluorophores (NIR998, NIR1028, NIR980, NIR1030, and NIR1028-S) based on aza-boron-dipyrromethene (aza-BODIPY) dyes with donor-acceptor-donor structures have been rationally designed and synthesized by harnessing the steric relaxation effect and intramolecular photoinduced electron transfer (IPET). These fluorophores exhibit an intense range of NIR-II emission, large Stokes shift (≥ 100 nm), excellent photothermal conversion performance, and superior stability against photobleaching. Among the NIR-II fluorophores, NIR998 possesses better NIR-II emission and photothermal conversion performance. NIR998 nanoparticles (NIR998 NPs) can be encapsulated by liposomes. NIR998 NPs show superior stability in the presence of light, heat, and reactive oxygen nitrogen species than that of indocyanine green NPs, as well as a higher photothermal conversion ability (η = 50.5%) compared to other photothermal agents. Finally, under the guidance of photothermal imaging, NIR998 NPs have been proven to effectively eliminate tumors via their excellent photothermal conversion performance while presenting negligible cytotoxicity. CONCLUSIONS: Utilizing IPET and the steric relaxation effect can effectively induce NIR-II emission of aza-BODIPY dyes. Stable NIR998 NPs have excellent photothermal conversion performance and negligible dark cytotoxicity, so they have the potential to act as photothermal agents in biological applications.


Subject(s)
Boron Compounds/therapeutic use , Fluorescent Dyes/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/therapy , Photothermal Therapy/methods , Animals , Boron Compounds/analysis , Boron Compounds/pharmacokinetics , Cell Line, Tumor , Female , Fluorescent Dyes/analysis , Fluorescent Dyes/pharmacokinetics , Humans , Infrared Rays , Mice , Nanoparticles/analysis , Neoplasms/diagnostic imaging , Theranostic Nanomedicine , Thermography
20.
Mikrochim Acta ; 188(9): 291, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34363101

ABSTRACT

A sensitive and turn-on fluorescence nanoprobe based on core-shell Ag@Au nanoparticles (Ag@AuNPs) as a fluorescence receptor and red emissive graphene quantum dots (GQDs) as a donor was fabricated. They were conjugated together through π-π stacking between the GQDs and single-strand DNA modified at the Ag@AuNPs surface. The absorption spectrum of the receptor significantly overlapped with the donor emission spectrum, leading to a strong Förster resonance energy transfer (FRET) and thus a dramatic quenching. The sensing mechanism relies on fluorescence recovery following DNA cleavage by •OH produced from Fenton-like reaction between the peroxidase-like Ag nanocore and H2O2. The red emissive feature (Ex/Em, 520 nm/560 nm) provides low background in physiological samples. The •OH production, great spectrum overlapping, and red emission together contributes to good sensitivity and living cell imaging capability. The fluorescence assay (intensity at 560 nm) achieves a low detection limit of 0.49 µM H2O2 and a wide linear range from 5 to 200 µM, superior to most of the reported fluorescent probes. The RSD value for 100 µM H2O2 was 1.4%. The nanoprobe exhibits excellent anti-interferences and shows low cytotoxicity. The recovery of 100 µM standard H2O2 in a cancer cell lysate was 85.8%. Most satisfactorily, it can realize monitoring and imaging H2O2 in living cells. This study not only presents a sensitive H2O2 probe but also provides a platform for detecting other types of reactive oxygen species.


Subject(s)
Fluorescent Dyes/therapeutic use , Gold/chemistry , Graphite/chemistry , Hydrogen Peroxide/chemistry , Metal Nanoparticles/chemistry , Quantum Dots/chemistry , Silver/chemistry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL