Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 65(24): 16679-16694, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36480920

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved cancer treatment that requires a photosensitizer (PS), light, and molecular oxygen─a combination which produces reactive oxygen species (ROS) that can induce cancer cell death. To enhance the efficacy of PDT, dual-targeted strategies have been explored where two photosensitizers are administered and localize to different subcellular organelles. To date, a single small-molecule conjugate for dual-targeted PDT with light-controlled nuclear localization has not been achieved. We designed a probe composed of a DNA-binding PS (Br-DAPI) and a photosensitizing photocage (WinterGreen). Illumination with 480 nm light removes WinterGreen from the conjugate and produces singlet oxygen mainly in the cytosol, while Br-DAPI localizes to nuclei, binds DNA, and produces ROS using one- or two-photon illumination. We observe synergistic photocytotoxicity in MCF7 breast cancer cells, and a reduction in size of three-dimensional (3D) tumor spheroids, demonstrating that nuclear/cytosolic photosensitization using a single agent can enhance PDT efficacy.


Subject(s)
Neoplasms , Photochemotherapy , Humans , Photosensitizing Agents/chemistry , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , DNA , Cell Line, Tumor
2.
Chem Commun (Camb) ; 58(78): 10929-10932, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36065979

ABSTRACT

Carboxylesterase 2 (CES2) has crucial roles in both xenobiotic metabolism and formation of pathogenic states including cancer. Thus, it is highly critical to monitor intracellular CES2 activity in living cancer cells. Here, we report a CES2 activatable phenoxy 1,2-dioxetane based chemiluminescent agent (CL-CES2). The probe exhibited a selective turn-on response in the presence of CES2 enzyme and enabled detection of CES2 activity in three different cancer cells that possess varying enzyme concentrations with high signal to noise ratios. In contrast no signal was obtained with CES1, an isoform of CES2 enzyme. CL-CES2 marks the first ever example of a CES2-responsive chemiluminescent luminophore and holds a great potential in further understanding the roles of CES2 activity in tumorogenesis.


Subject(s)
Carboxylic Ester Hydrolases , Neoplasms , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Protein Isoforms , Xenobiotics
4.
ACS Chem Biol ; 17(5): 1082-1091, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35394740

ABSTRACT

Reactive oxygen species (e.g., singlet oxygen) are the primary cytotoxic agents used in the clinically approved technique photodynamic therapy (PDT). Although singlet oxygen has high potential to effectively kill tumor cells, its production via light excitation of a photosensitizer has been limited by the penetration depth and delivery of light in tissue. To produce singlet oxygen without light excitation, we describe the use of Schaap's chemiluminescent scaffold comprising an adamantylidene-dioxetane motif. Functionalizing this scaffold with a photosensitizer, Erythrosin B, resulted in spontaneous chemiluminescence resonance energy transfer (CRET) leading to the production of singlet oxygen. We show that this compound is cell permeable and that the singlet oxygen produced via CRET is remarkably efficient in killing cancer cells at low micromolar concentrations. Moreover, we demonstrate that protection of the phenol on the chemiluminescent scaffold with a nitroreductase-responsive trigger group allows for cancer-selective dark dynamic cell death. Here, we present the concept of dark dynamic therapy using a small cell-permeable molecule capable of producing the effects of PDT in cells, without light.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Energy Transfer , Erythrosine , Luminescence , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Singlet Oxygen
5.
ACS Infect Dis ; 7(11): 3052-3061, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34617443

ABSTRACT

Antimicrobial photodynamic therapy (APDT) employs a photosensitizer, light, and molecular oxygen to treat infectious diseases via oxidative damage, with a low likelihood for the development of resistance. For optimal APDT efficacy, photosensitizers with cationic charges that can permeate bacteria cells and bind intracellular targets are desired to not limit oxidative damage to the outer bacterial structure. Here we report the application of brominated DAPI (Br-DAPI), a water-soluble, DNA-binding photosensitizer for the eradication of both Gram-negative and Gram-positive bacteria (as demonstrated on N99 Escherichia coli and Bacillus subtilis, respectively). We observe intracellular uptake of Br-DAPI, ROS-mediated bacterial cell death via one- and two-photon excitation, and selective photocytotoxicity of bacteria over mammalian cells. Photocytotoxicity of both N99 E. coli and B. subtilis occurred at submicromolar concentrations (IC50 = 0.2-0.4 µM) and low light doses (5 min irradiation times, 4.5 J cm-2 dose), making it superior to commonly employed APDT phenothiazinium photosensitizers such as methylene blue. Given its high potency and two-photon excitability, Br-DAPI is a promising novel photosensitizer for in vivo APDT applications.


Subject(s)
Escherichia coli , Photosensitizing Agents , Animals , Bacteria , DNA , Light , Photosensitizing Agents/pharmacology , Staphylococcus aureus , Water
6.
Chemistry ; 26(12): 2713-2718, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-31814180

ABSTRACT

Human NAD(P)H: Quinone Oxidoreductase 1 (hNQO1) is an attractive enzyme for cancer therapeutics due to its significant overexpression in tumors compared to healthy tissues. Its unique catalytic mechanism involving the two-electron reduction of quinone-based compounds has made it a useful target to exploit in the design of hNQO1 fluorescent chemosensors and hNQO1-activatable-prodrugs. In this work, hNQO1 is exploited for an optical therapeutic. The probe uses the photosensitizer, phenalenone, which is initially quenched via photo-induced electron transfer by the attached quinone. Native phenalenone is liberated in the presence of hNQO1 resulting in the production of cytotoxic singlet oxygen upon irradiation. hNQO1-mediated activation in A549 lung cancer cells containing high levels of hNQO1 induces a dose-dependent photo-cytotoxic response after irradiation. In contrast, no photo-cytotoxicity was observed in the normal lung cell line, MRC9. By targeting hNQO1, this scaffold can be used to enhance the cancer selectivity of photodynamic therapy.


Subject(s)
Antineoplastic Agents/chemistry , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD/metabolism , Phenalenes/chemistry , Photosensitizing Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , NAD(P)H Dehydrogenase (Quinone)/pharmacology , Phenalenes/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Quinones/metabolism , Singlet Oxygen/chemistry
7.
Chem Commun (Camb) ; 55(67): 9971-9974, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31367709

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved cancer treatment that uses light, oxygen and a photosensitizer to produce localized reactive oxygen species (ROS). Due to the short lifetime of ROS, the location of the photosensitizer in the cell is believed to be the key determinant governing the outcome of PDT. To explore the effect of direct association between a photosensitizer and DNA a well know DNA-binding dye, DAPI, was converted into a photosensitizer. Br-DAPI - unlike native DAPI - upon irradiation produces ROS. We demonstrate that the ROS are only effective in inducing dsDNA breaks when Br-DAPI is bound to DNA. In cancer cells (A549) Br-DAPI causes rapid light dependent cell death. This work supports the design of photosensitizers which bind with high affinity to the DNA of target cells for potentially more effective PDT.


Subject(s)
Bromine/chemistry , DNA/chemistry , Indoles/chemistry , Photosensitizing Agents/chemistry , A549 Cells , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage , Fluorescent Dyes/chemistry , Humans , Light , Photochemotherapy , Photosensitizing Agents/pharmacology , Proof of Concept Study , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...