Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
AAPS PharmSciTech ; 19(5): 2395-2406, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29869309

RESUMEN

Single chemotherapeutic agent like paclitaxel (PTX) has shown some limitations in anti-tumor treatment, such as undesirable side effects, multidrug resistance, and high toxicity. In order to reduce the toxicity of PTX and increase the anti-tumor effect, folate-modified amphiphilic and biodegradable biomaterial was developed to co-deliver PTX and metformin (MET) for exerting the synergistic effect. PTX was physically entrapped in the hydrophobic inner core of the amphiphilic block copolymer by a solvent evaporation method, whereas MET was chemically conjugated to the hydrophilic terminals of copolymer via a pH-sensitive cis-aconityl linkage (Cis). The in vitro release behaviors of the drugs were analyzed by high-performance liquid chromatography (HPLC), and the synergistic effect of the drugs was evaluated by a Q value method. Results showed that drug-loaded micelles with an average size about 100 nm were successfully constructed. In acidic environments, the chemically conjugated MET was rapidly released after the breakage of sensitive bond between drug and copolymer. In vitro anti-tumor studies demonstrated that MET and PTX had a synergistic effect and co-delivery micelles induced higher cytotoxicity and apoptosis against 4T1 breast cancer cells than free drugs. Furthermore, folate-targeted co-delivery micelles increased the cellular uptake of drugs and were found to be effective for the treatment of solid tumor in vivo. These findings indicated that co-delivery of MET and PTX through the polymeric micelles is a promising strategy for cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Ácido Fólico/administración & dosificación , Metformina/administración & dosificación , Paclitaxel/administración & dosificación , Carga Tumoral/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Femenino , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Metformina/química , Metformina/metabolismo , Ratones , Ratones Endogámicos BALB C , Micelas , Paclitaxel/química , Paclitaxel/metabolismo , Resultado del Tratamiento , Carga Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
Nat Commun ; 15(1): 7558, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39214974

RESUMEN

Hydrogen sulfide is essential in numerous physiological and pathological processes and has emerged as a promising cancer imaging and signaling molecule and a potentially versatile therapeutic agent. However, the endogenous levels of hydrogen sulfide remain insufficient to perform its biological functions, and thus, developing novel strategies that amplify hydrogen sulfide signals at lesion sites is of increasing interest. In this work, a nanoplatform (SNP) based on hydrogen sulfide-responsive self-immolative poly(thiocarbamate) with localized hydrogen sulfide signal amplification capability is developed to encapsulate a hydrogen sulfide-responsive fluorescent probe (e.g., hemicyanine dye; p-Cy) or an anticancer prodrug (e.g., doxorubicin; p-DOX) to form a nanoprobe (SNPp-Cy) or nanomedicine (SNPp-DOX) for cancer imaging and therapy, respectively. SNPp-Cy exhibits a low detection limit for hydrogen sulfide, enabling ultrasensitive detection of small (<2 mm) tumors in female mice. In addition, SNPp-DOX can effectively inhibit the growth of DOX-resistant human breast cancer xenograft, lung metastasis, and patient-derived xenograft tumors in female mice.


Asunto(s)
Doxorrubicina , Sulfuro de Hidrógeno , Sulfuro de Hidrógeno/metabolismo , Animales , Humanos , Femenino , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Doxorrubicina/administración & dosificación , Ratones , Línea Celular Tumoral , Colorantes Fluorescentes/química , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Profármacos/farmacología , Profármacos/uso terapéutico , Profármacos/química , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Ratones Endogámicos BALB C , Nanopartículas/química , Imagen Óptica/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
3.
Biomater Sci ; 12(14): 3649-3658, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38857014

RESUMEN

Despite cisplatin's pivotal role in clinically proven anticancer drugs, its application has been hampered by severe side effects and a grim prognosis. Herein, we devised a glutathione (GSH)-responsive nanoparticle (PFS-NP) that integrates a disulfide bond-based amphiphilic polyphenol (PP-SS-DA), a dopamine-modified cisplatin prodrug (Pt-OH) and iron ions (Fe3+) through coordination reactions between Fe3+ and phenols. After entering cells, the responsively released Pt-OH and disulfide bonds eliminate the intracellular GSH, in turn disrupting the redox homeostasis. Meanwhile, the activated cisplatin elevates the intracellular H2O2 level through cascade reactions. This is further utilized to produce highly toxic hydroxyl radicals (˙OH) catalyzed by the Fe3+-based Fenton reaction. Thus, the amplified oxidative stress leads to immunogenic cell death (ICD), promoting the maturation of dendritic cells (DCs) and ultimately activating the anti-tumor immune system. This innovative cisplatin prodrug nanoparticle approach offers a promising reference for minimizing side effects and optimizing the therapeutic effects of cisplatin-based drugs.


Asunto(s)
Antineoplásicos , Cisplatino , Profármacos , Cisplatino/farmacología , Cisplatino/química , Cisplatino/administración & dosificación , Profármacos/química , Profármacos/farmacología , Profármacos/administración & dosificación , Humanos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Animales , Ratones , Inmunoterapia/métodos , Nanopartículas/química , Nanopartículas/administración & dosificación , Células Dendríticas/efectos de los fármacos , Glutatión/química , Glutatión/metabolismo , Línea Celular Tumoral , Hierro/química , Portadores de Fármacos/química , Fenoles/química , Fenoles/farmacología , Fenoles/administración & dosificación
4.
Adv Mater ; 36(30): e2402322, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38718226

RESUMEN

Novel strategies to facilitate tumor-specific drug delivery and restore immune attacks remain challenging in overcoming the current limitations of chemoimmunotherapy. An antitumor chemoimmunotherapy system comprising bioorthogonal reaction-ready group tetrazine (TZ) modified with an anti-PD-L1 antibody (αPD-L1TZ) and TZ-activatable prodrug vinyl ether-doxorubicin (DOX-VE) for self-reinforced anti-tumor chemoimmunotherapy is proposed. The αPD-L1TZ effectively disrupts the PD-L1/PD-1 interaction and activates the DOX prodrug in situ through the bioorthogonal click reaction of TZ and VE. Conversely, the activated DOX upregulates PD-L1 on the surface of tumor cells, facilitating tumor accumulation of αPD-L1TZ and enhancing DOX-VE activation. Furthermore, the activated DOX-induced immunogenic cell death of tumor cells, substantially improving the response efficiency of αPD-L1 in an immune-suppressive tumor microenvironment. Thus, PD-L1 blocking and bioorthogonal in situ prodrug activation synergistically enhance the antitumor efficacy of the chemoimmunotherapy system. Therefore, the system significantly enhances αPD-L1 tumor accumulation and prodrug activation and induces a robust immunological memory effect to prevent tumor recurrence and metastasis. Thus, a feasible chemoimmunotherapy combination regimen is presented.


Asunto(s)
Antígeno B7-H1 , Doxorrubicina , Inmunoterapia , Profármacos , Profármacos/química , Profármacos/farmacología , Profármacos/uso terapéutico , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Inmunoterapia/métodos , Animales , Ratones , Línea Celular Tumoral , Humanos , Microambiente Tumoral/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
5.
Chem Commun (Camb) ; 59(21): 3142-3145, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36811610

RESUMEN

In this work, we have developed a tumor-specific self-accelerating prodrug activation nanosystem consisting of self-amplifying degradable polyprodrug PEG-TA-CA-DOX and encapsulated fluorescent prodrug BCyNH2, equipped with a reactive oxygen species dual-cycle amplification effect. Furthermore, activated CyNH2 is a therapeutic agent with potential to synergistically improve chemotherapy.


Asunto(s)
Nanopartículas , Profármacos , Profármacos/farmacología , Especies Reactivas de Oxígeno , Liberación de Fármacos , Sistemas de Liberación de Medicamentos , Doxorrubicina/farmacología , Línea Celular Tumoral
6.
Chem Commun (Camb) ; 59(26): 3878-3881, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36916644

RESUMEN

In this study, we combined low pH and cathepsin B dual-stimulus-triggered delivery carriers with a bioorthogonal reaction-activated prodrug to achieve regulated activation of the prodrug. A workable method for precise tumor therapy and imaging is provided by the bioorthogonal reaction, which activates the prodrug and fluorescent probe.


Asunto(s)
Profármacos , Profármacos/uso terapéutico , Colorantes Fluorescentes , Imagen Óptica/métodos
7.
Biomater Sci ; 10(15): 4228-4234, 2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-35758299

RESUMEN

The use of polyprodrugs, which bind drugs to polymer chains through responsive linkers, is a potential technique for cancer therapy; however, a lack of endogenous triggering factors limits drug activation in tumor tissue. Herein, we rationally created a reactive oxygen species (ROS)-sensitive polyprodrug (TSCA/DOX) with cascade amplification of triggering agents and drug activation by incorporating both an ROS signal amplifier (TACA) and a drug activation amplifier (SIPDOX) into a delivery system. Endogenous ROS as a triggering mechanism kicked off the initial circulation phase to increase intracellular ROS signals. Subsequently, the enhanced ROS initiated the second degradation step, allowing the polyprodrug SIPDOX to fracture spontaneously in a domino-like fashion, resulting in self-accelerated drug activation in tumor tissue. Therefore, the polyprodrug created in this study with cascade amplification of drug activation holds great promise for effective cancer treatment.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Humanos , Neoplasias/tratamiento farmacológico , Polímeros/metabolismo , Profármacos/farmacología , Profármacos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
8.
Acta Biomater ; 154: 97-107, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36210042

RESUMEN

The selective activation of stimuli-responsive polymers in the tumor microenvironment is a great concern to achieve intelligent cancer therapy, but most of them show inadequate response due to insufficient endogenous triggering agents. Herein, we rationally designed a reactive oxygen species (ROS)-responsive cinnamaldehyde (CA)-based poly(thioacetal), consisting of ROS-responsive thioacetal (TA) and ROS-generating agent CA, with self-amplified chain-shattering polymer degradation. The mechanism of self-amplified chain-shattering is that endogenous ROS as a triggering agent facilitates chain cleavage of TA with the release of CA, which in turn produces more ROS through mitochondrial dysfunction, resulting in an exponential polymer degradation cascade. The polymer can be further modified with anticancer drug doxorubicin (DOX) for cooperative amplification of oxidative stress and immunogenic cell death (ICD) of tumor cells, thereby boosting the effect of chemo-immunotherapy. The self-amplified chain-shattering polymer designed in this work holds great promise in developing stimuli-responsive polymers for efficient drug delivery. STATEMENT OF SIGNIFICANCE: This study presented an approach to utilize self-amplified chain-shattering cinnamaldehyde-based poly (thioacetal) as a drug delivery system to restrain tumor growth and boost chemo-immunotherapy. The endogenous ROS as a triggering agent initiates the chain cleavage with the release of CA, which in turn produces ROS through mitochondria dysfunction, resulting in an exponential polymer degradation cascade and rapid drug release.


Asunto(s)
Nanopartículas , Neoplasias , Polímeros de Estímulo Receptivo , Humanos , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Polímeros/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral , Microambiente Tumoral
9.
Biomaterials ; 289: 121803, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36150300

RESUMEN

Reactive oxygen species (ROS)-activated prodrugs can potentially improve the selectivity of chemotherapeutics. However, the inability to release sufficient drugs at tumor sites due to the paucity of ROS, which is required for prodrug activation usually limits the antitumor potency. Herein, a delivery nanosystem with self-amplifiable drug release pattern is constructed by encapsulating a tumor specificity ROS inducer NAD(P)H: quinone oxidoreductase-1 (NQO1)-responsive hemicyanine fluorescent dye (NCyNH2) in a ROS-responsive self-immolative polyprodrug nanoparticle for orchestrated oxidation-chemotherapy. In response to ROS stimulation, the self-immolative polyprodrug can degrade and release doxorubicin (DOX) through a domino-like fragmentation, which can impart advanced attributes of this nanosystem such as minimum cleavage events required and maximum cleavage speed for disintegration. Thus, the NCyNH2-loaded self-immolative polyprodrug nanoparticle (SIPN) could be dissociated in response to endogenous ROS, triggering the release of DOX and NCyNH2. Subsequently, the NCyNH2 could be activated by intratumoral overexpressed NQO1 to generate additional ROS, which further induces the amplifiable degradation of self-immolative polyprodrug to release sufficient drugs. The in vitro and in vivo studies consistently demonstrate that SIPN amplifies the drug release efficiency of ROS-responsive polyprodrug by specifically upregulating intratumoral ROS levels, resulting in significant antitumor efficacy with minimal side effects.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Colorantes Fluorescentes , Humanos , NAD/metabolismo , Neoplasias/tratamiento farmacológico , Profármacos/uso terapéutico , Quinonas , Especies Reactivas de Oxígeno/metabolismo
10.
Biomaterials ; 289: 121795, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36108580

RESUMEN

Although stimuli-responsive polymers have emerged as promising strategies for intelligent cancer therapy, limited polymer degradation and insufficient drug release remain a challenge. Here, we report a novel reactive oxygen species (ROS)-awakened self-amplifying degradable cinnamaldehyde (CA)-based poly(thioacetal) polymer. The polymer consists of ROS responsive thioacetal (TA) group and CA as the ROS generation agent. The self-amplified polymer degradation process is triggered by endogenous ROS-induced cleavage of the TA group to release CA. The CA released then promotes the generation of more ROS through mitochondrial dysfunction, resulting in amplified polymer degradation. More importantly, poly(thioacetal) itself can trigger immunogenic cell death (ICD) of the tumor cells and its side chains can be conjugated with indoleamine 2,3-dioxygenase 1 (IDO-1) inhibitor to reverse the immunosuppressive tumor microenvironment for synergistic cancer immunotherapy. The self-amplified degradable poly(thioacetal) developed in this work provides insights into the development of novel stimulus-responsive polymers for enhanced cancer immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Polímeros de Estímulo Receptivo , Acroleína/análogos & derivados , Línea Celular Tumoral , Humanos , Inmunoterapia , Indolamina-Pirrol 2,3,-Dioxigenasa/uso terapéutico , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Polímeros/química , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral
11.
ACS Nano ; 16(1): 721-735, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34978422

RESUMEN

Hypoxia, a common feature of most solid tumors, causes severe tumor resistance to chemotherapy and immunotherapy. Herein, a tumor-acidity and bioorthogonal chemistry-mediated on-site size transformation clustered nanosystem is designed to overcome hypoxic resistance and enhance chemoimmunotherapy. The nanosystem utilized the tumor-acidity responsive group poly(2-azepane ethyl methacrylate) with a rapid response rate and highly efficient bioorthogonal click chemistry to form large-sized aggregates in tumor tissue to enhance accumulation and retention. Subsequently, another tumor-acidity responsive group of the maleic acid amide with a slow response rate was cleaved allowing the aggregates to slowly dissociate into ultrasmall nanoparticles with better tumor penetration ability for the delivery of doxorubicin (DOX) and nitric oxide (NO) to a hypoxic tumor tissue. NO can reverse a hypoxia-induced DOX resistance and boost the antitumor immune response through a reprogrammed tumor immune microenvironment. This tumor-acidity and bioorthogonal chemistry-mediated on-site size transformation clustered nanosystem not only helps to counteract a hypoxia-induced chemoresistance and enhance antitumor immune responses but also provides a general drug delivery strategy for enhanced tumor accumulation and penetration.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Neoplasias/tratamiento farmacológico , Nanopartículas/uso terapéutico , Inmunoterapia , Microambiente Tumoral , Hipoxia/tratamiento farmacológico , Línea Celular Tumoral
12.
Chem Commun (Camb) ; 58(20): 3294-3297, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35175251

RESUMEN

In this work, we developed a ferroptosis self-catalyst, PTAF, exhibiting self-catalyzed ferroptosis for enhanced cancer therapy. Briefly, synergistic actions of self-catalyzed ˙OH accumulation and GPX4 indirect inactivation based on the establishment of the ROS self-catalytic loop effectively induced tumor ferroptosis, which provided a novel approach for enhanced tumor therapy.


Asunto(s)
Ferroptosis , Neoplasias , Catálisis , Línea Celular Tumoral , Humanos , Metalocenos , Polímeros , Especies Reactivas de Oxígeno
13.
J Control Release ; 348: 1004-1015, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35779654

RESUMEN

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.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Línea Celular Tumoral , Doxorrubicina , Quimioterapia Combinada , Humanos , Hipoxia , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Profármacos/uso terapéutico
14.
J Control Release ; 338: 307-315, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34454962

RESUMEN

Currently, stimulus-responsive nanomedicines are usually activated by a single cancer-associated biomarker and utilize different image/therapeutic agents for cancer imaging/therapy, which restricts the specificity of nanomedicine and complicates their design. Herein, we report a novel dual-locking theranostic nanoprobe (DL-P) based on near-infrared (NIR) hemicyanine CyNH2 with two orthogonal stimuli of cancer cell lysosomal pH (first "lock")- and lysosome-overexpressed cathepsin B (CTB, second "lock")-triggered NIR fluorescence turn-on and drug activation to improve the specificity of cancer imaging and therapy. The fluorescence of CyNH2 was initially quenched due to intramolecular charge transfer (ICT) but could be selectively activated under the dual-key stimulation of lysosomal pH and CTB to liberate CyNH2, resulting in strong NIR fluorescence turn-on for cancer imaging. Moreover, CyNH2 caused mitochondrial dysfunction to inhibit cancer cell proliferation in the absence of laser irradiation, which can be used in cancer therapy. Compared with previously reported probes that respond to a single stimulus, this dual-locking nanoprobe that is responsive to two orthogonal stimuli triggers with integrated imaging and therapy function in a single agent exhibits increased selectivity and specificity, which provides a prospective strategy for precise cancer imaging and therapy.


Asunto(s)
Neoplasias , Carbocianinas , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Imagen Óptica , Estudios Prospectivos , Nanomedicina Teranóstica
15.
Biomaterials ; 278: 121136, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34562835

RESUMEN

Combination therapy using multiple drugs with time-programmed administration is promising for enhanced cancer treatment. However, it is still challenging to achieve time-programmed drug release from a single nanocarrier. Here, dual polyprodrugs of hemicyanine dye (CyNH2) and doxorubicin (DOX) are developed to achieve time-programmed prodrug activation for synergistic cascade oxidation therapy and chemotherapy. The polyprodrug NPDOX/Cy, composed of CyNH2, is modified with a glutathione (GSH)-responsive disulfate group, while DOX is modified with a reactive oxygen species (ROS)-response thioketal (TK) group. Upon uptake by cancer cells overexpressing GSH, CyNH2 can be activated quickly and accumulate in the mitochondria to induce mitochondrial damage and ROS upregulation, thus achieving subsequent burst activation of DOX through the ROS-triggered cleavage of the TK linker. The early activation of CyNH2 makes the cancer cells more sensitive to subsequent DOX treatment for a synergistic effect of from oxidation therapy and chemotherapy. Therefore, the polyprodrug with time-programmed drug activation developed in this work provides a promising strategy for synergistic cancer therapy.


Asunto(s)
Nanopartículas , Profármacos , Línea Celular Tumoral , Doxorrubicina , Liberación de Fármacos
16.
Biomaterials ; 270: 120649, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33588139

RESUMEN

High intracellular glutathione (GSH) levels play an important role in multidrug resistance (MDR) in cancer cells. It remains challenging to develop a drug delivery system that is simultaneously capable of GSH depletion and drug activation for multidrug resistance reversal. Herein, we designed a polyprodrug (denoted as PSSD) based on poly(disulfide) conjugated with doxorubicin (DOX) on the polymer side chains that exhibits GSH depletion and cascade DOX activation for drug resistance reversal. The poly(disulfide) backbone with a high disulfide density depletes intracellular antioxidant GSH via the disulfide-thiol exchange reaction to disrupt intracellular redox homeostasis in cells. Simultaneously, DOX can be activated through a cascade reaction, and degradation of the poly(disulfide) backbone further facilitates its drug release. Therefore, poly(disulfide) can be used as a GSH scavenger to reverse MDR as well as a prodrug backbone to target high intracellular GSH levels in cancer cells, providing a general strategy for drug resistance reversal.


Asunto(s)
Resistencia a Antineoplásicos , Glutatión , Activación Metabólica , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Glutatión/metabolismo
17.
Biomaterials ; 276: 121005, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34252801

RESUMEN

Discovering new strategies to overcome multidrug resistance (MDR) is still urgently needed. MDR is associated with the overexpression of transmembrane efflux pumps, and adenosine triphosphate (ATP) is indispensable for its function. Herein, we developed a pH- and glutathione (GSH)-responsive amphiphilic poly(disulfide acetal) (PCS) containing cinnamaldehyde (CA) and disulfide groups that amplify oxidative stress for anticancer drug delivery and simultaneously overcome drug resistance in cancer cells. Reactive oxygen species (ROS)-generating CA and the disulfide groups to deplete GSH and synergize to amplify oxidative stress in cancer cells by oxidizing nicotinamide adenine dinucleotide with hydrogen (NADH) to nicotinamide adenine dinucleotide (NAD+). The production of ATP is preferentially inhibited, leading to the malfunction of efflux pumps due to the lack of ATP and making resistant cells more impressionable to anticancer drugs. The in vitro and in vivo experiments confirmed that PCS could induce amplified oxidative stress and efficiently overcome MDR in cancer cells. We believe that the polymer with amplified oxidative stress in cancer cells holds great promise in developing polymer-based drug delivery systems to reverse MDR for cancer therapy.


Asunto(s)
Acetales , Neoplasias , Disulfuros , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Neoplasias/tratamiento farmacológico , Estrés Oxidativo
18.
Adv Healthc Mater ; 10(21): e2101144, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34453773

RESUMEN

Theranostic prodrugs that can precisely monitor drug activation with synergistic therapeutic effects are highly desirable for personalized medicine. In this study, a theranostic heterodimeric prodrug, CyNH-SS-DOX, with synchronous and independent dual-channel fluorescence turn-on and dual-prodrug activation for synergistic cancer therapy is developed. A hemicyanine fluorescent drug, CyNH2 , with good therapeutic effects found in this work, is conjugated to doxorubicin (DOX) through a disulfide linker to form CyNH-SS-DOX. Before activation, both the fluorescence of DOX and CyNH2 are in the off state and the toxicity is low. In the presence of intracellular glutathione, both the fluorescence of DOX and CyNH2 at different channels are turned on. Meanwhile, DOX and CyNH2 are activated in a synergistic anticancer effect. It is believed that CyNH-SS-DOX is promising for monitoring prodrug activation in dual-fluorescence channels and for enhancing therapeutic efficacy with few side effects.


Asunto(s)
Neoplasias , Profármacos , Liberación de Fármacos , Fluorescencia , Humanos , Neoplasias/tratamiento farmacológico , Medicina de Precisión , Profármacos/farmacología
19.
Acta Biomater ; 135: 628-637, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34371167

RESUMEN

Subcellular organelle targeted imaging and therapy are of enormous interest in cancer theranostics. However, the lack of tumor-selective organelle targeting has compromised their efficacy and safety. In this work, we found that the near-infrared (NIR) fluorophore hemicyanine (CyNH2) can selectively target mitochondria with strong cytotoxicity through decreasing the mitochondrial membrane potential and increasing the intracellular reactive oxygen species (ROS) levels. A macrotheranostic probe (denoted as PLCy) based on conjugating CyNH2 with an acetylated lysine group was developed with masked fluorescence and cytotoxicity, which could both be unmasked through sequential activation by cancer cells overexpressing histone deacetylases (HDACs) and cathepsin L (CTSL) enzymes for selective cancer cell mitochondria-targeted imaging and therapy. In vitro and in vivo studies confirmed that the specific fluorescence turn-on and toxicity were restored in cancer cells and efficiently inhibited tumor growth. This macrotheranostic probe with sequential enzyme activation and mitochondrial targeting is expected to have promising applications in imaging-guided cancer therapy with high specificity and efficiency. STATEMENT OF SIGNIFICANCE: To improve the targeting efficiency and enhance the anti-cancer activities of macrotheranostic probe. We designed macrotheranostic probe PLCy that can be activated via sequential enzymes for selective tumor mitochondria targeting. More importantly, the activated CyNH2 can decrease the mitochondrial membrane potential and elevate the reactive oxygen species level in cancer cells without light irradiation, which can further induce apoptosis of tumor cells for chemotherapy. Therefore, the use of sequential enzyme activation and mitochondria targeting strategies in the context of enzymatic activation may provide a general strategy for organelle-targeted imaging and therapy with high specificity and efficiency.


Asunto(s)
Neoplasias , Línea Celular Tumoral , Fluorescencia , Colorantes Fluorescentes , Humanos , Mitocondrias , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno
20.
ACS Appl Mater Interfaces ; 12(6): 6933-6943, 2020 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-31951372

RESUMEN

The normoxic and hypoxic microenvironments in solid tumors cause cancer cells to show different sensitivities to various treatments. Therefore, it is essential to develop different therapeutic modalities based on the tumor microenvironment. In this study, we designed size-switchable nanoparticles with self-destruction and tumor penetration characteristics for site-specific phototherapy of cancer. This was achieved by photodynamic therapy in the perivascular normoxic microenvironment due to high local oxygen concentrations and photothermal therapy (PTT) in the hypoxic microenvironment, which are not in proximity to blood vessels due to a lack of effective approaches for heat transfer. In brief, a poly(amidoamine) dendrimer with photothermal agent indocyanine green (PAMAM-ICG) was conjugated to the amphiphilic polymer through a singlet oxygen-responsive thioketal linker and then loaded with photosensitizer chlorin e6 (Ce6) to construct a nanotherapy platform (denoted as SNPICG/Ce6). After intravenous injection, SNPICG/Ce6 was accumulated at the perivascular sites of the tumor. The singlet oxygen produced by Ce6 can ablate the tumor cells in the normoxic microenvironment and simultaneously cleave the thioketal linker, allowing the release of small PAMAM-ICGs with improved tumor penetration for PTT in the hypoxic microenvironment. This tailored site-specific phototherapy in normoxic and hypoxic microenvironments provides an effective strategy for cancer therapy.


Asunto(s)
Nanopartículas/química , Neoplasias/terapia , Fotoquimioterapia , Animales , Línea Celular Tumoral , Clorofilidas , Humanos , Verde de Indocianina/administración & dosificación , Verde de Indocianina/química , Ratones , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/química , Poliaminas/química , Porfirinas/administración & dosificación , Porfirinas/química , Oxígeno Singlete/administración & dosificación , Oxígeno Singlete/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA