Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(9): e2314620121, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38381784

RESUMO

Photon-controlled pyroptosis activation (PhotoPyro) is a promising technique for cancer immunotherapy due to its noninvasive nature, precise control, and ease of operation. Here, we report that biomolecular photoredox catalysis in cells might be an important mechanism underlying PhotoPyro. Our findings reveal that the photocatalyst lutetium texaphyrin (MLu) facilitates rapid and direct photoredox oxidation of nicotinamide adenine dinucleotide, nicotinamide adenine dinucleotide phosphate, and various amino acids, thereby triggering pyroptosis through the caspase 3/GSDME pathway. This mechanism is distinct from the well-established role of MLu as a photodynamic therapy sensitizer in cells. Two analogs of MLu, bearing different coordinated central metal cations, were also explored as controls. The first control, gadolinium texaphyrin (MGd), is a weak photocatalyst but generates reactive oxygen species (ROS) efficiently. The second control, manganese texaphyrin (MMn), is ineffective as both a photocatalyst and a ROS generator. Neither MGd nor MMn was found to trigger pyroptosis under the conditions where MLu was active. Even in the presence of a ROS scavenger, treating MDA-MB-231 cells with MLu at concentrations as low as 50 nM still allows for pyroptosis photo-activation. The present findings highlight how biomolecular photoredox catalysis could contribute to pyroptosis activation by mechanisms largely independent of ROS.


Assuntos
Metaloporfirinas , Piroptose , Espécies Reativas de Oxigênio/metabolismo
2.
Chem Rev ; 124(5): 2699-2804, 2024 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-38422393

RESUMO

The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.


Assuntos
Corantes Fluorescentes , Medicina de Precisão , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Fluorescência , Nanomedicina Teranóstica
3.
Proc Natl Acad Sci U S A ; 119(34): e2210504119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35969782

RESUMO

Elucidating the underlying photochemical mechanisms of action (MoA) of photodynamic therapy (PDT) may allow its efficacy to be improved and could set the stage for the development of new classes of PDT photosensitizers. Here, we provide evidence that "photoredox catalysis in cells," wherein key electron transport pathways are disrupted, could constitute a general MoA associated with PDT. Taking the cellular electron donor nicotinamide adenine dinucleotide as an example, we have found that well-known photosensitizers, such as Rose Bengal, BODIPY, phenoselenazinium, phthalocyanine, and porphyrin derivatives, are able to catalyze its conversion to NAD+. This MoA stands in contrast to conventional type I and type II photoactivation mechanisms involving electron and energy transfer, respectively. A newly designed molecular targeting photocatalyst (termed CatER) was designed to test the utility of this mechanism-based approach to photosensitizer development. Photoexcitation of CatER induces cell pyroptosis via the caspase 3/GSDME pathway. Specific epidermal growth factor receptor positive cancer cell recognition, high signal-to-background ratio tumor imaging (SBRTI = 12.2), and good tumor growth inhibition (TGI = 77.1%) are all hallmarks of CatER. CatER thus constitutes an effective near-infrared pyroptotic cell death photo-inducer. We believe the present results will provide the foundation for the synthesis of yet-improved phototherapeutic agents that incorporate photocatalytic chemistry into their molecular design.


Assuntos
Antineoplásicos , Neoplasias , Fotoquimioterapia , Fármacos Fotossensibilizantes , Antineoplásicos/farmacologia , Catálise , Linhagem Celular Tumoral , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia
4.
J Am Chem Soc ; 146(22): 15251-15263, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38780071

RESUMO

Glioblastoma (GBM) poses a significant therapeutic challenge due to its invasive nature and limited drug penetration through the blood-brain barrier (BBB). In response, here we present an innovative biomimetic approach involving the development of genetically engineered exosome nanocatalysts (Mn@Bi2Se3@RGE-Exos) for efficient GBM therapy via improving the BBB penetration and enzyme-like catalytic activities. Interestingly, a photothermally activatable multiple enzyme-like reactivity is observed in such a nanosystem. Upon NIR-II light irradiation, Mn@Bi2Se3@RGE-Exos are capable of converting hydrogen peroxide into hydroxyl radicals, oxygen, and superoxide radicals, providing a peroxidase (POD), oxidase (OXD), and catalase (CAT)-like nanocatalytic cascade. This consequently leads to strong oxidative stresses to damage GBM cells. In vitro, in vivo, and proteomic analysis further reveal the potential of Mn@Bi2Se3@RGE-Exos for the disruption of cellular homeostasis, enhancement of immunological response, and the induction of cancer cell ferroptosis, showcasing a great promise in anticancer efficacy against GBM with a favorable biosafety profile. Overall, the success of this study provides a feasible strategy for future design and clinical study of stimuli-responsive nanocatalytic medicine, especially in the context of challenging brain cancers like GBM.


Assuntos
Exossomos , Glioblastoma , Raios Infravermelhos , Fototerapia , Glioblastoma/tratamento farmacológico , Glioblastoma/terapia , Humanos , Exossomos/química , Exossomos/metabolismo , Animais , Fototerapia/métodos , Camundongos , Catálise , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Manganês/química , Manganês/farmacologia , Barreira Hematoencefálica/metabolismo
5.
Chem Soc Rev ; 52(3): 879-920, 2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36637396

RESUMO

Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.


Assuntos
Neoplasias , Fotoquimioterapia , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral
6.
J Am Chem Soc ; 145(11): 6007-6023, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36881923

RESUMO

Pyroptosis refers to the process of gasdermin-mediated lytic programmed cell death (PCD) characterized by the release of pro-inflammatory cytokines. Our knowledge of pyroptosis has expanded beyond the cellular level and now includes extracellular responses. In recent years, pyroptosis has attracted considerable attention due to its potential to induce host immunity. For instance, at the 2022 International Medicinal Chemistry of Natural Active Ligand Metal-Based Drugs (MCNALMD) conference, numerous researchers demonstrated an interest in photon-controlled pyroptosis activation ("PhotoPyro"), an emerging pyroptosis-engineered approach for activating systemic immunity via photoirradiation. Given this enthusiasm, we share in this Perspective our views on this emerging area and expound on how and why "PhotoPyro" could trigger antitumor immunity (i.e., turning so-called "cold" tumors "hot"). In doing so, we have tried to highlight cutting-edge breakthroughs in PhotoPyro while suggesting areas for future contributions. By providing insights into the current state of the art and serving as a resource for individuals interested in working in this area, it is hoped that this Perspective will set the stage for PhotoPyro to evolve into a broadly applicable cancer treatment strategy.


Assuntos
Neoplasias , Piroptose , Humanos , Apoptose , Imunidade , Neoplasias/tratamento farmacológico
7.
J Am Chem Soc ; 145(8): 4647-4658, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36745678

RESUMO

Cancer stem cells (CSCs) are associated with the invasion and metastatic relapse of various cancers. However, current cancer therapies are limited to targeting the bulk of primary tumor cells while remaining the CSCs untouched. Here, we report a new proton (H+) modulation approach to selectively eradicate CSCs via cutting off the H+ leaks on the inner mitochondrial membrane (IMM). Based on the fruit extract of Gardenia jasminoides, a multimodal molecule channel blocker with high biosafety, namely, Bo-Mt-Ge, is developed. Importantly, in this study, we successfully identify that mitochondrial uncoupling protein UCP2 is closely correlated with the stemness of CSCs, which may offer a new perspective for selective CSC drug discovery. Mechanistic studies show that Bo-Mt-Ge can specifically inhibit the UCP2 activities, decrease the H+ influx in the matrix, regulate the electrochemical gradient, and deplete the endogenous GSH, which synergistically constitute a unique MoA to active apoptotic CSC death. Intriguingly, Bo-Mt-Ge also counteracts the therapeutic resistance via a two-pronged tactic: drug efflux pump P-glycoprotein downregulation and antiapoptotic factor (e.g., Bcl-2) inhibition. With these merits, Bo-Mt-Ge proved to be one of the safest and most efficacious anti-CSC agents, with ca. 100-fold more potent than genipin alone in vitro and in vivo. This study offers new insights and promising solutions for future CSC therapies in the clinic.


Assuntos
Membranas Mitocondriais , Neoplasias , Humanos , Membranas Mitocondriais/metabolismo , Prótons , Neoplasias/patologia , Células-Tronco Neoplásicas/metabolismo
8.
Acc Chem Res ; 55(22): 3253-3264, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36323625

RESUMO

The advent of photochemical techniques has revolutionized the landscape of biology and medical sciences. Especially appealing in this context is photodynamic therapy (PDT), which is a photon-initiated treatment modality that uses cytotoxic reactive oxygen species (ROS) to kill malignant cells. In the past decade, PDT has risen to the forefront of cancer therapy. Its optical control enables noninvasive and spatiotemporal manipulation of the treatment process, and its photoactive nature allows unique patterns to avoid drug resistance to conventional chemotherapeutics. However, despite the impressive advances in this field, achieving widespread clinical adoption of PDT remains difficult. A major concern is that in the hostile tumor microenvironment, tumor cells are hypoxic, which hinders ROS generation during PDT action. To overcome this "Achilles' heel", current strategies focus primarily on the improvement of the intratumoral O2 perfusion, while clinical trials suggest that O2 enrichment may promote cancer cell proliferation and metastasis, thereby making FDA approval and clinical transformation of these paradigms challenging.In an effort to improve hypoxia photodynamic therapy (hPDT) in the clinic, we have explored "low to no O2-dependent" photochemical approaches over the years to combat hypoxia-induced resistance. In this Account, we present our contributions to this theme during the past 5 years, beginning with low O2-dependent approaches (e.g., type I superoxide radical (O2•-) generator, photodynamic O2-economizer, mitochondrial respiration inhibition, cellular self-protective pathway modulation, etc.) and progressing to O2-independent strategies (e.g., autoadaptive PDT/PTT complementary therapy, O2-independent artificial photoredox catalysis in cells). These studies have attracted tremendous attention. Particularly in the pioneering work of 2018, we presented the first demonstration that the O2•--mediated partial O2-recyclability mechanism can overcome PDT resistance ( J. Am. Chem. Soc. 2018, 140, 14851-14859). This launched an era of renewed interest in type I PDT, resulting in a plethora of new O2•- photogenerators developed by many groups around the world. Moreover, with the discovery of O2-independent photoredox reactions in living cells, artificial photoredox catalysis has emerged as a new field connecting photochemistry and biomedicine, stimulating the development of next-generation phototherapeutic tools ( J. Am. Chem. Soc. 2022, 144, 163-173). Our recent work also disclosed that "photoredox catalysis in cells" might be a general mechanism of action of PDT ( Proc. Natl. Acad. Sci. U.S.A. 2022, 119, e2210504119). These emergent concepts, molecular designs, photochemical mechanisms, and applications in cancer diagnosis and therapeutics, as well as pros and cons, are discussed in depth in this Account. It is expected that our contributions to date will be of general use to researchers and inspire future efforts to identify more promising hPDT approaches that better meet the clinical needs of cancer therapy.


Assuntos
Neoplasias , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Hipóxia , Neoplasias/tratamento farmacológico , Microambiente Tumoral
9.
Inorg Chem ; 62(45): 18767-18778, 2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-37905835

RESUMO

The remarkable impact of photoredox catalytic chemistries has sparked a wave of innovation, opening doors to novel biotechnologies in the realm of catalytic antitumor therapy. Yet, the quest for novel photoredox catalysts (PCs) suitable for living systems, or the enhancement of catalytic efficacy in existing biocompatible PC systems, persists as a formidable challenge. Within this context, we introduce a readily applicable metal modulation strategy that significantly augments photoredox catalysis within living cells, exemplified by a set of metalloporphyrin complexes termed M-TCPPs (M = Zn, Mn, Ni, Co, Cu). Among these complexes, Zn-TCPP emerges as an exceptional catalyst, displaying remarkable photocatalytic activity in the oxidation of nicotinamide adenine dinucleotide (NADH), nicotinamide adenine dinucleotide phosphate (NADPH), and specific amino acids. Notably, comprehensive investigations reveal that Zn-TCPP's superior catalytic prowess primarily arises from the establishment of an efficient oxidative cycle for PC, in contrast to previously reported PCs engaged in reductive cycles. Moreover, theoretical calculations illuminate that amplified intersystem crossing rates and geometry alterations in Zn-TCPP contribute to its heightened photocatalytic performance. In vitro studies demonstrated that Zn-TCPP exhibits therapeutic potential and is found to be effective for photocatalytic antitumor therapy in both glioblastoma G98T cells and 3D multicellular spheroids. This study underscores the transformative role of "metal modulation" in advancing high-performance PCs for catalytic antitumor therapy, marking a significant stride toward the realization of this innovative therapeutic approach.


Assuntos
Metaloporfirinas , Metais , Metais/química , Metaloporfirinas/farmacologia , Oxirredução , Catálise
10.
Chem Soc Rev ; 51(19): 8201-8215, 2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-36069855

RESUMO

Light-based phototherapy has been developed for cancer treatment owing to its non-invasiveness and spatiotemporal control. Despite the unique merits of phototherapy, one critical disadvantage of light is its limited penetration depth, which restricts its application in cancer treatment. Although many researchers have developed various strategies to deliver light into deep-seated tumors with two-photon and near-infrared light irradiation, phototherapy encounters the peculiar limitations of light. In addition, high oxygen dependency is another limitation of photodynamic therapy to treat hypoxic tumors. To overcome the drawbacks of conventional treatments, various energy sources have been developed for cancer treatment. Generally, most energy sources, such as ultrasound, chemiluminescence, radiation, microwave, electricity, and magnetic field, are relatively free from the restraint of penetration depth. Combining other strategies or therapies with other energy-source-based therapies improves the strength and compensates for the weakness. This tutorial review focuses on recent advances in the diverse energy sources utilized in cancer treatment and their future perspectives.


Assuntos
Neoplasias , Fotoquimioterapia , Humanos , Luminescência , Neoplasias/tratamento farmacológico , Oxigênio , Fototerapia
11.
Angew Chem Int Ed Engl ; 62(32): e202307797, 2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37336786

RESUMO

Photoacoustic imaging (PAI), a state-of-the-art noninvasive in vivo imaging technique, has been widely used in clinical disease diagnosis. However, the design of high-performance PAI agents with three key characteristics, i.e., near-infrared (NIR) absorption (λabs >800 nm), intense PA signals, and excellent photostability, remains a challenging goal. Herein, we present a facile but effective approach for engineering PAI agents by amplifying intramolecular low-frequency vibrations and enhancing the push-pull effect. As a demonstration of this blended approach, we constructed a PAI agent (BDP1-NEt2 ) based on the boron-dipyrromethene (BODIPY) scaffold. Compared with indocyanine green (ICG, an FDA-approved organic dye widely utilized in PAI studies; λabs =788 nm), BDP1-NEt2 exhibited a UV/Vis-NIR spectrum peaked at 825 nm, superior in vivo PA signal intensity and outstanding stability to offer improved tumor diagnostics. We believe this work provides a promising strategy to develop the next generation of PAI agents.


Assuntos
Técnicas Fotoacústicas , Técnicas Fotoacústicas/métodos , Vibração , Verde de Indocianina , Corantes , Diagnóstico por Imagem
12.
J Am Chem Soc ; 144(25): 11326-11337, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35708298

RESUMO

Pyroptosis, a newly characterized form of immunogenic cell death, is attracting increasing attention as a promising approach to cancer immunotherapy. However, biocompatible strategies to activate pyroptosis remain rare. Here, we show that a photocatalytic superoxide radical (O2-•) generator, NI-TA, triggers pyroptosis in cancer cells. NI-TA was designed to take advantage of an intramolecular triplet-ground state splitting energy modulation approach. Detailed studies revealed that the pyroptosis triggered by NI-TA under conditions of photoexcitation proceeds through a caspase-3/gasdermin E (GSDME) pathway rather than via canonical processes involving caspase-1/gasdermin-D (GSDMD). NI-TA was found to function via a partial-O2-recycling mode of action and to trigger cell pyroptosis and provide for effective cancer cell ablation even under conditions of hypoxia (≤2% O2). In the case of T47D 3D multicellular spheroids, good antitumor efficiency and stemness inhibition are achieved. This work highlights how photocatalytic chemistry may be leveraged to develop effective pyroptosis-inducing agents.


Assuntos
Neoplasias , Piroptose , Caspase 1/metabolismo , Caspase 1/farmacologia , Superóxidos
13.
J Am Chem Soc ; 144(1): 163-173, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34963281

RESUMO

The transformational effect of photoredox catalytic chemistries has inspired new opportunities, enabling us to interrogate nature in ways that are not possible otherwise and to unveil new biotechnologies in therapy and diagnosis. However, the deployment of artificial photoredox catalysis in living systems remains challenging, mired by the off-target risk and safety concerns of photocatalyst toxicity. Here, we present an appealing approach, namely conditionally activatable photoredox catalysis (ConAPC), and as a proof of concept design the first ConAPC architecture (Se-NO2) based upon classic self-immolative chemistry, in which the inherent photocatalytic properties can be temporarily caged while the species becomes active only at the tumor sites via sensing to specific biomarkers. Such a masking strategy allows a spatial-temporal control of photoresponsivity in vitro and in vivo. In particular, for ConAPC design, a new biologically benign metal-free photocatalyst (Se-NH2), which is able to initiate NIR photoredox catalysis to manipulate the cellular electron pool in an O2-independent mechanism of action, is identified. With this unique strategy, potent tumor-specific targeting photocatalytic eradication (TGI: 95%) is obtained in a mouse model. Impressively, favorable features such as high-resolution tumor recognition (SBR: 33.6) and excellent biocompatibility and safety are also achieved. This work therefore offers a new possibility for chemists to leverage artificial photocatalytic reactions toward the development of facile and intelligent photocatalytic theranostics.


Assuntos
Processos Fotoquímicos
14.
J Am Chem Soc ; 144(37): 16799-16807, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36070479

RESUMO

Ultrasound-triggered remote control of biomolecular functions in cells provides unique advantages for us to interrogate nature. However, strategies to design therapeutic ultrasound-responsive functional molecules remain elusive, and rare ultrasound-cleavable chemical bonds have been developed to date. Herein, therapeutic ultrasound (1 MHz)-induced scission of urea bonds for drug release is demonstrated for the first time. Such a transformation has been verified to be initiated by hydroxyl radicals generated in the interior of cavitation bubbles, occurring specifically at the cavitation bubble-liquid interface. A series of urea-bond-containing prodrugs based on methylene blue (MB), namely MBUs, are designed. Upon sonication with low-intensity therapeutic ultrasound, the urea bonds linked with primary amines can be selectively cleaved, and free MB is released in a physiologically relevant environment, accompanied by recovered absorbance, fluorescence, and photosensitivity. Moreover, an FDA-approved alkylating agent (i.e., melphalan) bearing urea bond is also developed (MBU-Mel), successfully achieving ultrasound-triggered drug release in deep-seated cancer cells (mimic with 1 cm pigskin), showing the scalability of our ultrasound-responsive molecule platform in bioactive molecules release. This may set the starting point for therapeutic ultrasound-induced drug release, making a forward step in "sonopharmacology".


Assuntos
Pró-Fármacos , Ureia , Alquilantes , Aminas , Melfalan , Azul de Metileno/farmacologia
15.
Angew Chem Int Ed Engl ; 61(17): e202110832, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35142018

RESUMO

Despite extensive efforts to realize effective photodynamic therapy (PDT), there is still a lack of therapeutic approaches concisely structured to mitigate the major obstacles of PDT in clinical applications. Herein, we report a molecular strategy exploiting ascorbate chemistry to enhance the efficacy of PDT in cancer cells overexpressing glucose transporter 1 (GLUT1). AA-EtNBS, a 5-O-substituted ascorbate-photosensitizer (PS) conjugate, undergoes a reversible structural conversion of the ascorbate moiety in the presence of reactive oxygen species (ROS) and glutathione (GSH), thereby promoting its uptake in GLUT1-overexpressed KM12C colon cancer cells and perturbing tumor redox homeostasis, respectively. Due to the unique pro-oxidant role of ascorbate in tumor environments, AA-EtNBS effectively sensitized KM12C cancer cells prior to PS-mediated generation of superoxide radicals under near-infrared (NIR) illumination. AA-EtNBS successfully exhibited GLUT1-targeted synergistic therapeutic efficacy during PDT both in vitro and in vivo. Therefore, this study outlines a promising strategy employing ascorbate both as a targeting unit for GLUT1-overexpressed cancer cells and redox homeostasis destruction agent, thereby enhancing therapeutic responses towards anticancer treatment when used in conjunction with conventional PDT.


Assuntos
Neoplasias , Fotoquimioterapia , Ácido Ascórbico/farmacologia , Linhagem Celular Tumoral , Transportador de Glucose Tipo 1 , Glutationa/metabolismo , Humanos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
16.
J Am Chem Soc ; 142(3): 1510-1517, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31880443

RESUMO

It remains a considerable challenge to realize complete tumor suppression and avoid tumor regrowth by rational design of photosensitizers (PSs) to improve their photon utilization. In this Article, we provide a molecular design (Icy-NBF) based on the oxygen-content-regulated deactivation process of excited states. In the presence of overexpressed nitroreductase in hypoxic cancer cells, Icy-NBF is reduced and converted into a molecule with the same skeleton (Icy-NH2), in which the deactivation of the PS under 808 nm light irradiation proceeds via a different pathway: the excited states deactivation pathway of Icy-NBF involves radiative transition and energy transfer between Icy-NBF and O2; as for Icy-NH2, the deactivation pathway is attributed to non-radiative relaxation. By varying the O2 concentration in tumor cells, the therapeutic mechanism of Icy-NBF under 808 nm light irradiation can be switched between photodynamic and photothermal therapies, which maximizes the advantages of phototherapies with no tumor regrowth. Our study provides help in designing of smart PSs with improvement of photon utilization for efficient tumor photoablation.


Assuntos
Oxigênio/química , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Terapia Fototérmica/métodos , Linhagem Celular Tumoral , Humanos , Cinética
17.
J Am Chem Soc ; 142(11): 5380-5388, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-32105455

RESUMO

Tumor hypoxia has proven to be the major bottleneck of photodynamic therapy (PDT) to clinical transformation. Different from traditional O2 delivery approaches, here we describe an innovative binary photodynamic O2-economizer (PDOE) tactic to reverse hypoxia-driven resistance by designing a superoxide radical (O2•-) generator targeting mitochondria respiration, termed SORgenTAM. This PDOE system is able to block intracellular O2 consumption and down-regulate HIF-1α expression, which successfully rescues cancer cells from becoming hypoxic and relieves the intrinsic hypoxia burden of tumors in vivo, thereby sparing sufficient endogenous O2 for the PDT process. Photosensitization mechanism studies demonstrate that SORgenTAM has an ideal intersystem crossing rate and triplet excited state lifetime for generating O2•- through type-I photochemistry, and the generated O2•- can further trigger a biocascade to reduce the PDT's demand for O2 in an O2-recycble manner. Furthermore, SORgenTAM also serves to activate the AMPK metabolism signaling pathway to inhibit cell repair and promote cell death. Consequently, using this two-step O2-economical strategy, under relatively low light dose irradiation, excellent therapeutic responses toward hypoxic tumors are achieved. This study offers a conceptual while practical paradigm for overcoming the pitfalls of phototherapeutics.


Assuntos
Neoplasias/tratamento farmacológico , Fenotiazinas/uso terapêutico , Fármacos Fotossensibilizantes/uso terapêutico , Hipóxia Tumoral/efeitos dos fármacos , Animais , Respiração Celular/efeitos dos fármacos , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Luz , Células MCF-7 , Camundongos Endogâmicos BALB C , Mitocôndrias/efeitos dos fármacos , Fenotiazinas/síntese química , Fenotiazinas/efeitos da radiação , Fotoquimioterapia , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/efeitos da radiação , Superóxidos/metabolismo
18.
Small ; 16(18): e1907677, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32307872

RESUMO

Targeting is one of the most important strategies for enhancing the efficacy of cancer photothermal therapy (PTT) and reducing damage to surrounding normal tissues. Compared with the traditional targeting approaches, the active targeting of breast cancer cells in PTT using chemotherapeutic drugs, such as tamoxifen (TAM), in combination with single-molecule photothermal photosensitizers has superior selectivity and therapeutic effects. However, single-molecule drug-targeting photosensitizers for improved PTT efficacy are not widely reported. Accordingly, herein, a near-infrared induced small-molecule photothermal photosensitizer (CyT) is developed that actively targets the estrogen receptors (ERs) of breast cancer cells as well as targets mitochondria by structure-inherent targeting. Cell uptake and cytotoxicity studies using different types of cells show that CyT enhances the efficiency of TAM-based PTT by targeting ER-overexpressing breast cancer cells and selectively killing them. In vivo experiments demonstrate that CyT can be used as a photothermal agent for fluorescence imaging-guided PTT. More importantly, the intravenous injection of CyT results in better targeting and efficiency of tumor inhibition compared with that achieved with the TAM-free control molecule Cy. Thus, the study presents an excellent small-molecule photothermal agent for breast cancer therapy with potential clinical application prospects.


Assuntos
Neoplasias da Mama , Sistemas de Liberação de Medicamentos , Hipertermia Induzida , Fármacos Fotossensibilizantes , Terapia Fototérmica , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia
19.
J Am Chem Soc ; 141(6): 2695-2702, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30652866

RESUMO

Strong oxygen dependence, poor tumor targeting, and limited treatment depth have been considered as the "Achilles' heels" facing the clinical usage of photodynamic therapy (PDT). Different from common approaches, here, we propose an innovative tactic by using photon-initiated dyad cationic superoxide radical (O2-•) generator (ENBOS) featuring "0 + 1 > 1" amplification effect to simultaneously overcome these drawbacks. In particular, by taking advantage of the Förster resonance energy transfer theory, the energy donor successfully endows ENBOS with significantly enhanced NIR absorbance and photon utility, which in turn lead to ENBOS more easily activated and generating more O2-• in deep tissues, that thus dramatically intensifies the type I PDT against hypoxic deep tumors. Moreover, benefiting from the dyad cationic feature, ENBOS achieves superior "structure-inherent targeting" abilities with the signal-to-background ratio as high as 25.2 at 48 h post intravenous injection, offering opportunities for accurate imaging-guided tumor treatment. Meanwhile, the intratumoral accumulation and retention performance are also markedly improved (>120 h). On the basis of these unique merits, ENBOS selectively inhibits the deep-seated hypoxic tumor proliferation at a low light-dose irradiation. Therefore, this delicate design may open new horizons and cause a paradigm change for PDT in future cancer therapy.


Assuntos
Fotoquimioterapia , Superóxidos/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , Camundongos , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação
20.
J Am Chem Soc ; 140(46): 15820-15826, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30380856

RESUMO

Structure-inherent targeting (SIT) agents are of particular importance for clinical precision medicine; however, there still exists a great lack of SIT phototheranostics for simultaneous cancer diagnosis and targeted photodynamic therapy (PDT). Herein, for the first time, we propose a "one-for-all" strategy by using the Förster resonance energy transfer (FRET) mechanism to construct such omnipotent SIT phototheranostics. Of note, this novel tactic can not only endow conventional sensitizers with highly effective native tumor-targeting potency but also simultaneously improve their photosensitization activities, resulting in dramatically boosted therapeutic index. After intravenous injection of the prepared SIT theranostic, the neoplastic sites are distinctly "lighted up" and distinguished from neighboring tissues, showing a near-infrared signal-to-background ratio value as high as 12.5. More importantly, benefiting from the FRET effect, markedly amplified light-harvesting ability and 1O2 production are demonstrated. Better still, other favorable features are also simultaneously achieved, including specific mitochondria anchoring, augmented cellular uptake (>13-fold), as well as ideal biocompatibility, all of which allow orders-of-magnitude promotion in anticancer efficiency both in vitro and in vivo. We believe this one-for-all SIT platform will provide a new idea for future cancer precision therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Desenho de Fármacos , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Transferência Ressonante de Energia de Fluorescência , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA