Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Biomacromolecules ; 25(10): 6635-6644, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39163639

ABSTRACT

Chemical reaction systems that can occur via multiple pathways in a controllable fashion are highly attractive for advanced materials applications and biological research. In this report, we introduce a bioorthogonal reaction manifold based on a chalcone pyrene (CPyr) moiety that can undergo either red-shifted photoreversible [2 + 2] cycloaddition or thiol-Michael addition click reaction. By coupling the CPyr to a water-soluble poly(ethylene glycol) end group, we demonstrate the efficient polymer dimerization and cleavage by blue light (λ = 450 nm) and UV light (λ = 340 nm), respectively. In the absence of light, CPyr rapidly reacts with thiols in aqueous environments, enabling fast and efficient polymer end-group functionalization. The chemical reaction manifold was further employed in polymer cross-linking for the preparation of hydrogels whose stiffness and morphology can be modulated by different photonic fields or the addition of a thiol cross-linker. The photoreversible cycloaddition and thiol-Michael addition click reaction can be used in conjunction for spatial and temporal conjugation of a streptavidin protein. Both cross-linking conditions are nontoxic to various cell lines, highlighting their potential in biomaterials applications.


Subject(s)
Click Chemistry , Cycloaddition Reaction , Hydrogels , Polyethylene Glycols , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Pyrenes/chemistry , Humans , Light , Sulfhydryl Compounds/chemistry , Cross-Linking Reagents/chemistry , Biocompatible Materials/chemistry
2.
J Am Chem Soc ; 146(33): 23376-23386, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39115375

ABSTRACT

Photocontrolled deprotection of specific functional groups has garnered significant interest over the past two decades. Notably, the selective deprotection of distinct groups based on wavelength has emerged as a prominent focus in recent research. The achievement of this objective has primarily involved the utilization of linker-based bichromophoric systems and diverse cocktail mixtures of photoresponsive protecting groups (PRPGs), each responsive to varying wavelengths of light. Herein, we present the first wavelength-selective monochromophoric system based on a hydroxanthene moiety, enabling the wavelength-selective release of two distinct functionalities under 450 and 600 nm light, respectively. The mechanism of the wavelength-selective photodegradation was thoroughly investigated by 1H NMR, UV-vis, and fluorescence spectroscopy, suggesting a proton-coupled electron transfer mechanism in the first photorelease step and electron transfer based arylmethyl type of photorelease in the second step. The utility of the xanthene-based wavelength-selective PRPGs was demonstrated in the multistep degradation of microparticles and dual-color tuning of polymer chain architecture, thus opening an avenue to design advanced photoreactive wavelength-controlled systems for applications in soft matter materials.

3.
J Chem Inf Model ; 64(14): 5580-5589, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38982947

ABSTRACT

Targeted treatments for breast cancer that minimize harm to healthy cells are highly sought after. Our study explores the potentiality of riboflavin as a targeted anticancer compound that can be activated by light irradiation. Here, we integrated time-dependent density functional theory (TD-DFT) calculations and an in vitro study under visible light. The TD-DFT calculations revealed that the electronic charge transferred from the DNA base to riboflavin, with the most significant excitation peak occurring within the visible light range. Guided by these insights, an in vitro study was conducted on the breast cancer cell lines MCF-7 and MDA-MB-231. The results revealed substantial growth inhibition in these cell lines when exposed to riboflavin under visible light, with no such impact observed in the absence of light exposure. Interestingly, riboflavin exhibited no/minimal growth-inhibitory effects on the normal cell line L929, irrespective of light conditions. Moreover, through EtBr displacement (DNA-EtBr) and the TUNEL assay, it has been illustrated that, upon exposure to visible light, riboflavin can intercalate within DNA and induce DNA damage. In conclusion, under visible light conditions, riboflavin emerges as a promising candidate with a selective and effective potent anticancer agent against breast cancer while exerting a minimal influence on regular cellular activity.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , DNA Damage , Density Functional Theory , Light , Riboflavin , Riboflavin/pharmacology , Riboflavin/chemistry , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Female , Cell Proliferation/drug effects , DNA , Models, Molecular
4.
Org Lett ; 26(29): 6236-6240, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39004831

ABSTRACT

We introduce a molecular engineering strategy for wavelength-selective photorelease of alcohols and carboxylic acids, employing a dynamic photocage featuring an (E)-3-(6-acetyl-2-hydroxynaphthalen-1-yl)acrylate moiety. Initially activated by visible light (λ ≥ 410 nm), it releases alcohol, generating a second photocage. Subsequent exposure to shorter wavelengths (λ ≤ 365 nm) selectively releases carboxylic acid, albeit at a reduced rate.

6.
ACS Appl Mater Interfaces ; 16(17): 21486-21497, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38640485

ABSTRACT

The combined effects of twisted intramolecular charge transfer (TICT) and aggregation-induced emission (AIE) phenomena have demonstrated a significant influence on excited-state chemistry. These combined TICT and AIE features have been extensively utilized to enhance photodynamic and photothermal therapy. Herein, we demonstrated the synergistic capabilities of TICT and AIE phenomena in the design of the photoremovable protecting group (PRPG), namely, NMe2-Napy-BF2. This innovative PRPG incorporates TICT and AIE characteristics, resulting in four remarkable properties: (i) red-shifted absorption wavelength, (ii) strong near-infrared (NIR) emission, (iii) viscosity-sensitive emission property, and (iv) accelerated photorelease rate. Inspired by these intriguing attributes, we developed a nanodrug delivery system (nano-DDS) using our PRPG for cancer treatment. In vitro studies showed that our nano-DDS manifested effective cellular internalization, specific staining of cancer cells, high-resolution confocal imaging of cancerous cells in the NIR region, and controlled release of the anticancer drug chlorambucil upon exposure to light, leading to cancer cell eradication. Most notably, our nano-DDS exhibited a substantially increased two-photon (TP) absorption cross section (435 GM), exhibiting its potential for in vivo applications. This development holds promise for significant advancements in cancer treatment strategies.


Subject(s)
Naphthyridines , Photons , Humans , Naphthyridines/chemistry , Naphthyridines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chlorambucil/chemistry , Chlorambucil/pharmacology , Photochemotherapy , Cell Line, Tumor , Cell Survival/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Nanoparticles/chemistry
7.
ACS Appl Bio Mater ; 6(10): 4372-4382, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37791981

ABSTRACT

Lipid droplets (LDs) are dynamic complex organelles involved in various physiological processes, and their number and activity are linked to multiple diseases, including cancer. In this study, we have developed LD-specific near-infrared (NIR) light-responsive nano-drug delivery systems (DDSs) based on chalcone derivatives for cancer treatment. The reported nano-DDSs localized inside the cancer microenvironment of LDs, and upon exposure to light, they delivered the anticancer drug valproic acid in a spatiotemporally controlled manner. The developed systems, namely, 2'-hydroxyacetophenone-dimethylaminobenzaldehyde-valproic (HA-DAB-VPA) and 2'-hydroxyacetophenone-diphenylaminobenzaldehyde-valproic (HA-DPB-VPA) ester conjugates, required only two simple synthetic steps. Our reported DDSs exhibited interesting properties such as excited-state intramolecular proton transfer (ESIPT) and aggregation-induced emission (AIE) phenomena, which provided advantages such as AIE-initiated photorelease and ESIPT-enhanced rate of photorelease upon exposure to one- or two-photon light. Further, colocalization studies of the nano-DDSs by employing two cancerous cell lines (MCF-7 cell line and CT-26 cell line) and one normal cell line (HEK cell line) revealed LD concentration-dependent enhanced fluorescence intensity. Furthermore, systematic investigations of both the nano-DDSs in the presence and absence of oleic acid inside the cells revealed that nano-DDS HA-DPB-VPA accumulated more selectively in the LDs. This unique selectivity by the nano-DDS HA-DPB-VPA toward the LDs is due to the hydrophobic nature of the diphenylaminobenzaldehyde (mimicking the LD core), which significantly leads to the aggregation and ESIPT (at 90% volume of fw, ΦF = 20.4% and in oleic acid ΦF = 24.6%), respectively. Significantly, we used this as a light-triggered anticancer drug delivery model to take advantage of the high selectivity and accumulation of the nano-DDS HA-DPB-VPA inside the LDs. Hence, these findings give a prototype for designing drug delivery models for monitoring LD-related intracellular activities and significantly triggering the release of LD-specific drugs in the biological field.


Subject(s)
Antineoplastic Agents , Lipid Droplets , Lipid Droplets/chemistry , Oleic Acid/analysis , Antineoplastic Agents/chemistry , Drug Delivery Systems
8.
Chem Sci ; 14(36): 9872-9884, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37736623

ABSTRACT

Installing proton-coupled electron transfer (PCET) in Ir-complexes is indeed a newly explored phenomenon, offering high quantum efficiency and tunable photophysics; however, the prospects for its application in various fields, including interrogating biological systems, are quite open and exciting. Herein, we developed various organelle-targeted Ir(iii)-complexes by leveraging the photoinduced PCET process to see the opportunities in phototherapeutic application and investigate the underlying mechanisms of action (MOAs). We diversified the ligands' nature and also incorporated a H-bonded benzimidazole-phenol (BIP) moiety with π-conjugated ancillary ligands in Ir(iii) to study the excited-state intramolecular proton transfer (ESIPT) process for tuning dual emission bands and to tempt excited-state PCET. These visible or two-photon-NIR light activatable Ir-catalysts generate reactive hydroxyl radicals (˙OH) and simultaneously oxidize electron donating biomolecules (1,4-dihydronicotinamide adenine dinucleotide or glutathione) to disrupt redox homeostasis, downregulate the GPX4 enzyme, and amplify oxidative stress and lipid peroxide (LPO) accumulation. Our homogeneous photocatalytic platform efficiently triggers organelle dysfunction mediated by a Fenton-like pathway with spatiotemporal control upon illumination to evoke ferroptosis poised with the synergistic action of apoptosis in a hypoxic environment leading to cell death. Ir2 is the most efficient photochemotherapy agent among others, which provided profound cytophototoxicity to 4T1 and MCF-7 cancerous cells and inhibited solid hypoxic tumor growth in vitro and in vivo.

9.
ACS Appl Mater Interfaces ; 15(27): 32099-32109, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37386863

ABSTRACT

The regulation of the cell cycle has recently opened up a new research perspective for cancer treatment. So far, no effort has been made for temporal control of cell cycles using a photocleavable linker. Presented herein is the first report of regulation of disrupted cell cycles through the temporal release of a well-known cell cycle regulator α-lipoic acid (ALA), enabled by a newly designed NIR-active quinoxaline-based photoremovable protecting group (PRPG). The suitable quinoxaline-based photocage of ALA (tetraphenylethelene conjugated) has been formulated as fluorescent organic nanoparticles (FONs) and used effectively as a nano-DDS (drug delivery system) for better solubility and cellular internalization. Fascinatingly, the enhanced TP (two-photon) absorption cross section of the nano-DDS (503 GM) signifies its utility for biological applications. Using green light, we have successfully controlled the time span of cell cycles and cell growth of skin melanoma cell lines (B16F10) by the temporal release of ALA. Further, in silico studies and PDH activity assay supported the observed regulatory behavior of our nano-DDS with respect to photoirradiation. Overall, this approach expands the research path toward a futuristic photocontrolled toolbox for cell cycle regulation.


Subject(s)
Nanoparticles , Prodrugs , Thioctic Acid , Nanoparticle Drug Delivery System , Quinoxalines/pharmacology , Drug Delivery Systems/methods , Cell Cycle
10.
J Mater Chem B ; 11(23): 5131-5141, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37234001

ABSTRACT

Hydrogen sulfide (H2S) is an important gasotransmitter that plays a significant role in the regulation of various physiological activities. The therapeutic effect of H2S is highly concentration-dependent and has recently been recognized for wound healing applications. Until now, the reported H2S delivery systems for wound healing applications have been focused on polymer-coated cargo systems for the encapsulation of H2S donors that are based just on endogenous stimuli-responsive systems such as pH or glutathione. These delivery systems lack spatio-temporal control and can cause premature H2S release depending on the wound microenvironment. In this regard, polymer-coated light-activated gasotransmitter donors provide a promising and efficient means of delivering high spatial and temporal control along with localized delivery. Hence, for the first time, we developed a ß-carboline photocage-based H2S donor (BCS) and formulated it into two photo-controlled H2S delivery systems: (i) Pluronic-coated nanoparticles loaded with BCS (Plu@BCS nano); and (ii) a hydrogel platform impregnated with BCS (Plu@BCS hydrogel). We investigated the mechanism of photo-release and the photo-regulated H2S release profile from the BCS photocage. We found that the Plu@BCS nano and Plu@BCS hydrogel systems were stable and did not release H2S without light treatment. Interestingly, external light manipulation, such as changing the irradiation wavelength, time, and location, regulate the release of H2S precisely. Biological studies (in vitro) suggest that the Pluronic coating on the BCS photocage makes the donor highly biocompatible and desirable for biological applications.


Subject(s)
Gasotransmitters , Hydrogen Sulfide , Nanoparticles , Poloxamer , Polymers , Hydrogels , Carbolines
11.
J Med Chem ; 66(6): 3732-3745, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36913722

ABSTRACT

Targeted release of bioactive molecules for therapeutic purposes is a key area in the biomedical field that is growing quickly, where bioactive molecules are released passively or actively from drug delivery systems (DDSs) or bioactive donors. In the past decade, researchers have identified light as one of the prime stimuli that can implement the efficient spatiotemporally targeted delivery of drugs or gaseous molecules with minimal cytotoxicity and a real-time monitoring ability. This perspective emphasizes recent advances in the photophysical properties of ESIPT- (excited-state intramolecular proton transfer), AIE- (aggregation-induced emission), and AIE + ESIPT-attributed light-activated delivery systems or donors. The three major sections of this perspective describe the distinctive features of DDSs and donors concerning their design, synthesis, photophysical and photochemical properties, and in vitro and in vivo studies demonstrating their relevance as carrier molecules for releasing cancer drugs and gaseous molecules in the biological system.


Subject(s)
Antineoplastic Agents , Drug Delivery Systems , Antineoplastic Agents/pharmacology , Protons
12.
Chem Commun (Camb) ; 59(9): 1177-1180, 2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36628583

ABSTRACT

Light-activated H2S donors have attracted considerable interest in understanding the physiological role and clinical potential of H2S, as their release is highly localized and controlled. Herein, we have evolved a new HPQ chromophore-based fluorescent organic nanosystem localized in a target area that tolerates oxidative stress and precisely releases H2S under one- and two-photon irradiation with real-time monitoring capability. The two-photon absorption cross-section of this new phototrigger was calculated to be 283 GM at 720 nm. H2S photorelease was also demonstrated in vitro on the MDA-MB-468 cell line in the presence of excess ROS. Our developed H2S nanoprodrug can be applied to living systems to release the H2S-gasotransmitter under laser irradiation in a phototherapeutic window.


Subject(s)
Fluorescent Dyes , Hydrogen Sulfide , Humans , HeLa Cells , Hydrogen Sulfide/metabolism , Photons , Oxidative Stress
13.
J Control Release ; 348: 1004-1015, 2022 08.
Article in English | MEDLINE | ID: mdl-35779654

ABSTRACT

Hypoxia-induced intratumoral heterogeneity poses a major challenge in tumor therapy due to the varying susceptibility to chemotherapy. Moreover, the spatial distribution patterns of hypoxic and normoxic tissues makes conventional combination therapy less effective. In this study, a tumor-acidity and bioorthogonal chemistry mediated in situ size transformable nanocarrier (NP@DOXDBCO plus iCPPAN3) was developed to spatially deliver two combinational chemotherapeutic drugs (doxorubicin (DOX) and PR104A) to combat hypoxia-induced intratumoral heterogeneity. DOX is highly toxic to tumor cells in normoxia state but less toxic in hypoxia state due to the hypoxia-induced chemoresistance. Meanwhile, PR104A is a hypoxia-activated prodrug has less toxic in normoxia state. Two nanocarriers, NP@DOXDBCO and iCPPAN3, can cross-link near the blood vessel extravasation sites through tumor acidity responsive bioorthogonal click chemistry to enhance the retention of DOX in tumor normoxia. Moreover, PR104A conjugated to the small-sized dendritic polyamidoamine (PAMAM) released under tumor acidity can penetrate deep tumor tissues for hypoxic tumor cell killing. Our study has demonstrated that this site-specific combination chemotherapy is better than the traditional combination chemotherapy. Therefore, spatial specific delivery of combinational therapeutics via in situ size transformable nanocarrier addresses the challenges of hypoxia induced intratumoral heterogeneity and provides insights into the combination therapy.


Subject(s)
Nanoparticles , Neoplasms , Prodrugs , Cell Line, Tumor , Doxorubicin , Drug Therapy, Combination , Humans , Hypoxia , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/therapeutic use
14.
Chem Soc Rev ; 51(6): 2255-2312, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35229836

ABSTRACT

The generation of azide radical (N3˙) occurs from its precursors primarily via a single electron transfer (SET) process or homolytic cleavage by chemical methods or advanced photoredox/electrochemical methods. This in situ generated transient open-shell species has unique characteristic features that set its reactivity. In the past, the azide radical was widely used for various studies in radiation chemistry as a 1e- oxidant of biologically important molecules, but now it is being exploited for synthetic applications based on its addition and intermolecular hydrogen atom transfer (HAT) abilities. Due to the significant role of nitrogen-containing molecules in synthesis, drug discovery, biological, and material sciences, the direct addition onto unsaturated bonds for the simultaneous construction of C-N bond with other (C-X) bonds are indeed worth highlighting. Moreover, the ability to generate O- or C-centered radicals by N3˙ via electron transfer (ET) and intermolecular HAT processes is also well documented. The purpose of controlling the reactivity of this short-lived intermediate in organic transformations drives us to survey: (i) the history of azide radical and its structural properties (thermodynamic, spectroscopic, etc.), (ii) chemical reactivities and kinetics, (iii) methods to produce N3˙ from various precursors, (iv) several significant azide radical-mediated transformations in the field of functionalization with unsaturated bonds, C-H functionalization via HAT, tandem, and multicomponent reaction with a critical analysis of underlying mechanistic approaches and outcomes, (v) concept of taming the reactivity of azide radicals for potential opportunities, in this review.


Subject(s)
Azides , Hydrogen , Electron Transport , Hydrogen/chemistry , Nitrogen , Thermodynamics
15.
ACS Appl Bio Mater ; 5(3): 1202-1209, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35148052

ABSTRACT

Developing green or red light-activated drug delivery systems (DDSs) for cancer treatment is highly desirable. Herein, we have reported a green light-responsive single component-based organic fluorescence nano-DDS by simply anchoring 2-hydroxy-6-naphthacyl (phototrigger) on both sides of the 1,5-diaminonaphthalene (DAN) chromophore. This green light (λ ≥ 500 nm)-activated DDS released two equivalents of the anticancer drug (valproic acid) in a spatio-temporally controlled manner. Our photoresponsive DDS [DAN-bis(HO-Naph-VPA)] exhibited interesting properties such as excited-state intramolecular proton transfer (ESIPT) accompanied with aggregation-induced emission (AIE) phenomena. AIE initiated the photorelease, and ESIPT enhanced the rate of the photorelease. Further, in vitro studies revealed that our green light-activated nano-DDS exhibited good cytocompatibility, excellent cellular internalization, and effective cancer cell killing ability.


Subject(s)
Antineoplastic Agents , Nanoparticle Drug Delivery System , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Fluorescence , Protons
16.
Chem Commun (Camb) ; 58(16): 2754-2757, 2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35128556

ABSTRACT

A phenothiazine-based photoremovable protecting group (PRPG) for single and dual release of carboxylic acids was developed. The change in the oxidation state of the sulfur atom of the phenothiazine PRPG resulted in singlet oxygen generation, rather than photorelease. The difference in the photochemistry between oxygen-free and oxygen-functionalized phenothiazine was investigated and supported by DFT calculations.

17.
ACS Appl Mater Interfaces ; 14(4): 4862-4870, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35049266

ABSTRACT

In recent times, organelle-targeted drug delivery systems have gained tremendous attention due to the site-specific delivery of active drug molecules, resulting in enhanced bioefficacy. In this context, a phototriggered drug delivery system (DDS) for releasing an active molecule is superior, as it provides spatial and temporal control over the release. So far, a near-infrared (NIR) light-responsive organelle-targeted DDS has not yet been developed. Hence, we introduced a two-photon NIR light-responsive lysosome-targeted "AIE + ESIPT" active single-component DDS based on the naphthalene chromophore. The two-photon absorption cross section of our DDS is 142 GM at 850 nm. The DDS was converted into pure organic nanoparticles for biological applications. Our nano-DDS is capable of selective targeting, AIE luminogenic imaging, and drug release within the lysosome. In vitro studies using cancerous cell lines showed that our single-component photoresponsive nanocarrier exhibited enhanced cytotoxicity and real-time monitoring ability of drug release.


Subject(s)
Biocompatible Materials/chemistry , Nanoparticles/chemistry , Naphthalenes/chemistry , Photons , Biocompatible Materials/chemical synthesis , Biocompatible Materials/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Delivery Systems , Humans , Infrared Rays , Materials Testing , Molecular Structure , Naphthalenes/pharmacology , Particle Size , Surface Properties , Time Factors
18.
J Med Chem ; 65(3): 1822-1834, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35019659

ABSTRACT

Herein, we report three new metal-free, photochemically active single, dual, and combinatorial CORMs (photoCORMs) based on a carbazole-fused 1,3-dioxol-2-one moiety which released one equivalent of CO, two equivalent of CO, and a combination of one equivalent of each CO and anticancer drug upon one- and two-photon excitation, respectively. The photoCORMs exhibited good cellular uptake and real-time monitoring ability of CO uncaging by a color change approach in cancerous B16F10 cells. Interestingly, the cytotoxicity assay on B16F10 cells indicated that the dual photoCORM has increased anticancer activity over the single and combinatorial photoCORMs upon irradiation. Our results also showed that CO could accelerate the effectiveness of the well-known anticancer drug (chlorambucil). Finally, the in vivo evaluation of the dual photoCORM on an established murine melanoma tumor (C57BL/6J mouse model) manifested a significant regression of tumor volume and led to significant improvement (>50%) in the overall survivability.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Carbon Monoxide/metabolism , Dioxoles/therapeutic use , Melanoma/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Apoptosis/drug effects , Carbazoles/chemical synthesis , Carbazoles/radiation effects , Cell Line, Tumor , Dioxoles/chemical synthesis , Dioxoles/radiation effects , Female , Infrared Rays , Mice, Inbred C57BL , Photons
20.
Org Lett ; 23(6): 2308-2313, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33689383

ABSTRACT

We demonstrated for the first time without any chemical modification the two-photon absorption (TPA) cross-section can be enhanced and red-shifted to the near-infrared (NIR) region by the ground-state proton-transfer (GSPT) process. Using GSPT, we developed a simple binol-based aggregation-induced emission (AIE)-fluorogenic phototrigger having a large two-photon uncaging cross-section in the "phototherapeutic window". As a proof of concept, we showed our phototrigger for the release of two different anticancer drugs in the NIR region.

SELECTION OF CITATIONS
SEARCH DETAIL