Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Am Chem Soc ; 146(1): 1185-1195, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38148611

RESUMO

Patients treated with Pt-based anticancer drugs (PtII) often experience severe side effects and are susceptible to cancer recurrence due to the limited bioavailability of PtII and tumor-induced immunosuppression. The exposure of phosphatidylserine on the cell's outer surface induced by PtII results in profound immunosuppression through the binding of phosphatidylserine to its receptors on immune cells. Here, we report a novel approach for enhanced cancer chemoimmunotherapy, where a novel nuclear-targeting lipid PtIV prodrug amphiphile was used to deliver a small interfering RNA (siXkr8) to simultaneously amplify Pt-DNA adducts and reduce the level of exposure of phosphatidylserine. This drug delivery vehicle is engineered by integrating the PtIV prodrug with self-assembly performance and siXkr8 into a lipid nanoparticle, which shows tumor accumulation, cancer cell nucleus targeting, and activatable in a reduced microenvironment. It is demonstrated that nuclear-targeting lipid PtIV prodrug increases the DNA cross-linking, resulting in increased Pt-DNA adduct formation. The synergistic effects of the PtIV prodrug and siXkr8 contribute to the improvement of the tumor immune microenvironment. Consequently, the increased Pt-DNA adducts and immunogenicity effectively inhibit primary tumor growth and prevent tumor recurrence. These results underscore the potential of utilizing the nuclear-targeting lipid PtIV prodrug amphiphile to enhance Pt-DNA adduct formation and employing siXkr8 to alleviate immunosuppression during chemotherapy.


Assuntos
Antineoplásicos , Neoplasias , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Adutos de DNA , Fosfatidilserinas , RNA Interferente Pequeno , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , RNA de Cadeia Dupla , Linhagem Celular Tumoral , Cisplatino , Microambiente Tumoral
2.
Mol Pharm ; 21(9): 4297-4311, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39106330

RESUMO

Inhibition of autophagy increases the sensitivity of tumor cells to radiotherapy and chemotherapy and improves the therapeutic effect on tumors. Recently, photodynamic therapy (PDT) combined with chemotherapy has been proven to further improve the efficiency of cancer treatment. As such, combining autophagy inhibition with PDT and chemotherapy may represent a potentially effective new strategy for cancer treatment. However, currently widely studied autophagy inhibitors inevitably produce various toxic side effects due to their inherent pharmacological activity. To overcome this constraint, in this study, we designed an ideal multifunctional upconversion nanoplatform, UCNP-Ce6-EPI@mPPA + NIR (MUCEN). Control, UCNP-EPI@mPPA (MUE), UCNP-EPI@mPPA + NIR (MUEN), Ce6-EPI@mPPA (MCE), Ce6-EPI@mPPA + NIR (MCEN), and UCNP-Ce6-EPI@mPPA (MUCE) groups were set up separately as controls. Based on a combination of autophagy inhibition and PDT, the average particle size of MUCEN was 197 nm, which can simultaneously achieve the double encapsulation of chlorine e6 (Ce6) and epirubicin (EPI). In vitro tests revealed that MUCE was efficiently endocytosed by 4T1 cells under near-infrared light irradiation. Further, in vivo tests revealed that MUCE dramatically inhibited tumor growth. Immunohistochemistry results indicated that MUCE efficiently increased the expression of autophagy inhibitors p62 and LC3 in tumor tissues. The synergistic effect of autophagy inhibition and PDT with MUCE exhibited superior tumor suppression, providing an innovative approach to cancer treatment.


Assuntos
Autofagia , Clorofilídeos , Camundongos Endogâmicos BALB C , Nanopartículas , Fotoquimioterapia , Fotoquimioterapia/métodos , Autofagia/efeitos dos fármacos , Animais , Camundongos , Nanopartículas/química , Linhagem Celular Tumoral , Humanos , Feminino , Epirubicina/farmacologia , Epirubicina/química , Porfirinas/química , Porfirinas/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Nus , Antineoplásicos/farmacologia , Antineoplásicos/química
3.
Angew Chem Int Ed Engl ; 61(20): e202201486, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35212437

RESUMO

The development of PtIV prodrugs which are selectively reduced within cancerous cells into their PtII therapeutically active species has received increasing attention within the last decade. Despite recent research progress, the majority of investigated compounds are excited using ultraviolet or blue light. As the light penetration depth is low at these wavelengths, the treatment of deep-seated or large tumors is limited. To overcome this limitation, herein, the example of PtIV -functionalized nanoparticles that could be excited within the NIR region at 808 nm is reported. The polymer backbone which can self-assemble into nanoparticles was functionalized with PtIV complexes for chemotherapy, photosensitizers for photodynamic immunotherapy, and nucleus/cancer-targeting peptides. Upon irradiation, the PtIV center is reduced to PtII and the axially coordinated ligands are released, presenting a multimodal treatment. While selectively accumulating in tumorous tissue, the nanoparticles demonstrated the ability to eradicate a triple-negative breast cancer tumor inside a mouse model.


Assuntos
Nanopartículas , Fotoquimioterapia , Pró-Fármacos , Animais , Linhagem Celular Tumoral , Imunoterapia , Camundongos , Nanopartículas/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Pró-Fármacos/química
4.
Bioorg Chem ; 88: 102916, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31026719

RESUMO

Both c-Met and VEGFR-2 were important targets for cancer therapies. In order to develop reversible and non-covalent c-Met and VEGFR-2 dual inhibitors, a series of [1,4]dioxino[2,3-f]quinazoline derivatives were designed and synthesized. The enzyme assay demonstrated that most target compounds had inhibition potency on both c-Met and VEGFR-2 with IC50 values in nanomolar range especially compounds 7m and 7k. Based on further cell proliferation assay in vitro, compound 7k showed significantly anti-tumor activity in vivo on a hepatocellular carcinoma (MHCC97H cells) xenograft mouse model. We docked the compound 7m with c-Met and VEGFR-2 kinases, and interpreted the SAR of these analogues. All results indicated that the target compounds were dual inhibitors of c-Met and VEGFR-2 kinases that held promising potential in cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Dioxanos/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Quinazolinas/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dioxanos/síntese química , Dioxanos/metabolismo , Feminino , Humanos , Ligação de Hidrogênio , Camundongos SCID , Simulação de Acoplamento Molecular , Estrutura Molecular , Proteínas Proto-Oncogênicas c-met/metabolismo , Quinazolinas/síntese química , Quinazolinas/metabolismo , Relação Estrutura-Atividade , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Angew Chem Int Ed Engl ; 58(40): 14224-14228, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31389144

RESUMO

The targeted delivery of chemotherapeutic drugs is a major challenge in the clinical treatment of cancer. Herein, we constructed a multifunctional DNA nanoplatform as a versatile carrier of the highly potent platinum-based DNA intercalator, 56MESS. In our rational design, 56MESS was efficiently loaded into the double-bundle DNA tetrahedron through intercalation with the DNA duplex. With the integration of a nanobody that both targets and blocks epidermal growth factor receptor (EGFR), the DNA nanocarriers exhibit excellent selectivity for cells with elevated EGFR expression (a common biomarker related to tumor formation) and combined tumor therapy without obvious systemic toxicity. This DNA-based platinum-drug delivery system provides a promising strategy for the treatment of tumors.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , DNA/química , Sistemas de Liberação de Medicamentos , Nanoestruturas/química , Compostos Organoplatínicos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Compostos Organoplatínicos/síntese química , Compostos Organoplatínicos/química
6.
Adv Healthc Mater ; 13(7): e2302746, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37988194

RESUMO

Chemotherapy remains the primary treatment method for osteosarcoma after surgery. However, the lack of selectivity of chemotherapy for osteosarcoma leads to unpredictable therapeutic effects, undesirable side effects, and drug resistance. A platinum(IV) (PtIV ) prodrug amphiphile (ALN-PtIV -Lipo) covalently bound to alendronate (ALN) and a lipid tail is designed to overcome these limitations. ALN-PtIV -Lipo can self-assemble into PtIV lipid nanoparticles (APtIV ) for osteosarcoma targeting chemotherapy and bone destruction inhibition. It is demonstrated that APtIV achieved an eightfold increase in the eradication of osteosarcoma cells compared to cisplatin and threefold selective inhibition of osteosarcoma cells over breast cancer cells via APtIV in vitro. After intravenous injection, APtIV effectively accumulates at the osteosarcoma site in vivo, resulting in significantly suppressed primary osteosarcoma growth, and alleviation of bone destruction. Therefore, APtIV delivers a promising solution for enhanced chemotherapy targeting and bone destruction inhibition in osteosarcoma.


Assuntos
Antineoplásicos , Neoplasias Ósseas , Nanopartículas , Osteossarcoma , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Antineoplásicos/farmacologia , Alendronato/uso terapêutico , Cisplatino/uso terapêutico , Osteossarcoma/tratamento farmacológico , Linhagem Celular Tumoral
7.
J Control Release ; 373: 216-223, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39002797

RESUMO

Platinum(II)-based drugs (PtII), which hinder DNA replication, are the most widely used chemotherapeutics. However, current PtII drugs often miss their DNA targets, leading to severe side effects and drug resistance. To overcome this challenge, we developed a oxaliplatin-based platinum(IV) (PtIV) prodrug amphiphile (C16-OPtIV-R8K), integrating a long-chain hydrophobic lipid and a nucleus-targeting hydrophilic peptide (R8K). This design allows the prodrug to self-assemble into highly uniform lipid nanoparticles (NTPtIV) for enhanced targeting chemotherapy and immunotherapy. Subsequently, NTPtIV's bioactivity and effects were examined at diverse levels, encompassing cancer cells, 3D tumor spheres, and in vivo. Our in vitro studies show a 74% cancer cell nucleus localization of platinum drugs-3.6 times higher than that of oxaliplatin, achieving more than a ten-fold increase in eliminating drug-resistant cancer cells. In vivo, NTPtIV shows efficient tumor accumulation, leading to suppressed tumor growth of murine breast cancer. Moreover, NTPtIV recruited more CD4+ and CD8+ T cells and reduced CD4+ Foxp3+ Tregs to synergistically enhance targeted chemotherapy and immunotherapy. Overall, this strategy presents a promising advancement in nucleus-targeted cancer therapy, synergistically boosting the efficacy of chemotherapy and immunotherapy.


Assuntos
Antineoplásicos , Imunoterapia , Compostos Organoplatínicos , Oxaliplatina , Pró-Fármacos , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Antineoplásicos/química , Humanos , Imunoterapia/métodos , Feminino , Linhagem Celular Tumoral , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/farmacologia , Compostos Organoplatínicos/química , Oxaliplatina/administração & dosagem , Oxaliplatina/farmacologia , Núcleo Celular/metabolismo , Nanopartículas/química , Camundongos Endogâmicos BALB C , Camundongos , Lipídeos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia
8.
Adv Mater ; 36(30): e2402452, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38691849

RESUMO

The in vivo fate of chemotherapeutic drugs plays a vital role in understanding the therapeutic outcome, side effects, and the mechanism. However, the lack of imaging abilities of drugs, tedious labeling processes, and premature leakage of imaging agents result in loss of fidelity between the drugs and imaging signals. Herein, an amphiphilic polymer is created by copolymerization of a near-infrared-II (NIR-II) fluorophore tracer (T) and an anticancer Pt(IV) prodrug (D) of cisplatin in a hand-holding manner into one polymer chain for the first time. The obtained PolyplatinDT is capable of delivering the drugs and the fluorophores concomitantly at a precise D/T ratio, thereby resulting in tracking the platinum drugs and even readout of them in real-time via NIR-II imaging. PolyplatinDT can self-assemble into nanoparticles, referred to as NanoplatinDT. Furthermore, a caspase-3 cleavable peptide that serves as an apoptosis reporter is attached to NanoplatinDT, resulting in NanoplatinDTR that are capable of simultaneously tracking platinum drugs and evaluating the therapeutic efficacy. Overall, it is reported here the design of the first theranostic polymer with anticancer drugs, drug tracers, and drug efficacy reporters that can work in concert to provide insight into the drug fate and mechanism of action.


Assuntos
Antineoplásicos , Corantes Fluorescentes , Pró-Fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Humanos , Corantes Fluorescentes/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Cisplatino/química , Cisplatino/farmacologia , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Polímeros/química , Camundongos , Imagem Óptica , Apoptose/efeitos dos fármacos , Portadores de Fármacos/química
9.
Nanoscale ; 15(36): 14790-14799, 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37642471

RESUMO

Photodynamic therapy has been used as a treatment option for cancer; however, the existing TiO2 photosensitizer does not have the ability to specifically target cancer cells. This lack of selectivity reduces its effectiveness in overcoming cancer resistance. To improve photodynamic therapy outcomes, an innovative solution is proposed. In this study, we report on the compounding of a zwitterionic covalent organic polymer (COP) with a TiO2 photosensitizer for the first time. The aim is to overcome cancer cellular resistance. A one-pot synthetic strategy, which includes the construction of a porphyrin-based COP has been employed. This strategy has also been applied to the rapid preparation of anatase defective TiO2 (TiO2-x). To improve the hydrophilic and antifouling properties of the polymer, zwitterion L-cysteine has been conjugated with a porphyrin-based COP using a thiol-ene "click chemistry" reaction. The novel zwitterionic porphyrin-based COP has the ability to trigger biodegradation under the acid microenvironment due to the presence of acid-sensitive ß-thioether esters. When combined with TiO2-x, the resultant nanocomposite produces an enhanced photodynamic therapy effect for drug-resistant cancer cells under NIR laser irradiation. This is due to the strong mutual sensitization of zwitterionic porphyrin-based COP and TiO2-x. Importantly, the nanocomposite delivery system exhibits excellent cytocompatibility in the dark and has the potential to improve the accuracy of cancer diagnosis through fluorescence imaging. The results of this study demonstrate the potential application of this alternative nanocomposite delivery system for remote-controllable photodynamic therapy of tumors.


Assuntos
Nanocompostos , Neoplasias , Fotoquimioterapia , Porfirinas , Fármacos Fotossensibilizantes/farmacologia , Nanocompostos/uso terapêutico , Polímeros , Porfirinas/farmacologia , Neoplasias/tratamento farmacológico
10.
ACS Nano ; 17(23): 23223-23261, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38041800

RESUMO

Stimuli-responsive polymers can respond to internal stimuli, such as reactive oxygen species (ROS), glutathione (GSH), and pH, biological stimuli, such as enzymes, and external stimuli, such as lasers and ultrasound, etc., by changing their hydrophobicity/hydrophilicity, degradability, ionizability, etc., and thus have been widely used in biomedical applications. Due to the characteristics of the tumor microenvironment (TME), stimuli-responsive polymers that cater specifically to the TME have been extensively used to prepare smart nanovehicles for the targeted delivery of therapeutic and diagnostic agents to tumor tissues. Compared to conventional drug delivery nanosystems, TME-responsive nanosystems have many advantages, such as high sensitivity, broad applicability among different tumors, functional versatility, and improved biosafety. In recent years, a great deal of research has been devoted to engineering efficient stimuli-responsive polymeric nanosystems, and significant improvement has been made to both cancer diagnosis and therapy. In this review, we summarize some recent research advances involving the use of stimuli-responsive polymer nanocarriers in drug delivery, tumor imaging, therapy, and theranostics. Various chemical stimuli will be described in the context of stimuli-responsive nanosystems. Accordingly, the functional chemical groups responsible for the responsiveness and the strategies to incorporate these groups into the polymer will be discussed in detail. With the research on this topic expending at a fast pace, some innovative concepts, such as sequential and cascade drug release, NIR-II imaging, and multifunctional formulations, have emerged as popular strategies for enhanced performance, which will also be included here with up-to-date illustrations. We hope that this review will offer valuable insights for the selection and optimization of stimuli-responsive polymers to help accelerate their future applications in cancer diagnosis and treatment.


Assuntos
Neoplasias , Polímeros Responsivos a Estímulos , Humanos , Medicina de Precisão , Sistemas de Liberação de Medicamentos/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Polímeros/uso terapêutico , Microambiente Tumoral
11.
Acta Biomater ; 160: 198-210, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36792048

RESUMO

As a DNA damaging agent, oxaliplatin (OXA) can induce immunogenic cell death (ICD) in tumors to activate the immune system. However, the DNA damage induced by OXA is limited and the ICD effect is not strong enough to enhance anti-tumor efficacy. Here, we propose a strategy to maximize the ICD effect of OXA through the mild hyperthermia generated by nanoparticles with a platinum (IV) prodrug of OXA (Pt(IV)-C16) and a near-infrared-II (NIR-II) photothermal agent IR1061 upon the irradiation of NIR-II light. The mild hyperthermia (43 °C) holds advantages in two aspects: 1) increase the Pt-DNA cross-linking, leading to enhanced DNA damage and apoptosis; 2) induce stronger ICD effects for cancer immunotherapy. We demonstrated that, compared with OXA and photothermal therapy of IR1061 alone, these nanoparticles under NIR-II light irradiation can significantly improve the anti-cancer efficacy against triple-negative breast cancer 4T1 tumor. This new strategy provides an effective way to improve the therapeutic outcome of OXA. STATEMENT OF SIGNIFICANCE: OXA could induce immunogenic cell death (ICD) via stimulating immune responses by increasing tumor cell stress and death, which triggers tumor-specific immune responses to achieve immunotherapy. However, due to the insufficient Pt-DNA crosslinks, the ICD effect triggered by OXA cannot induce robust immune response. Mild hyperthermia has great potential to maximize the therapeutic outcome of oxaliplatin by increasing the Pt-DNA cross-linking to augment the immunoresponse for enhanced cancer immunotherapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Oxaliplatina/farmacologia , Morte Celular Imunogênica , Boratos , Neoplasias/tratamento farmacológico , Imunoterapia , DNA , Linhagem Celular Tumoral
12.
Anal Chim Acta ; 1279: 341779, 2023 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-37827677

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disorder that devastatingly affects people's lives. Accumulating evidence indicates that the pathological progression of AD is inseparably connected with hypochlorous acid (HClO). However, further exploring the biological function remains an open challenging due to a lack of effective tools to image HClO in AD brains. To this end, a ruthenium(II) luminescence probe, Ru-HClO, is developed for quantitative detection and visualization of HClO in nerve cells and AD brains. Ru-HClO shows quenched luminescence due to the PET process (excited electron transfer from Ru(II) center to diaminomaleonitrile) and the CN bond isomerization in the excited state. The HClO-triggered specific cleavage reaction with Ru-HClO cleaves the CN bond to form highly luminescent Ru-COOH. Ru-HClO shows rapid response speed, high sensitivity and selectivity, excellent biocompatibility, which makes the probe to be applied to semi-quantitative analysis of HClO in nerve cells and high-throughput screening of anti-AD drugs in the AD cell model. Moreover, using Ru-HClO as a probe, present work further validated that the elevated levels of HClO secretion were accompanied by the AD progressed. These findings may provide valuable results for figuring out the biological roles that HClO played in AD but also for accelerating anti-AD therapeutic discovery.


Assuntos
Doença de Alzheimer , Rutênio , Humanos , Luminescência , Ácido Hipocloroso/análise , Rutênio/química , Doença de Alzheimer/diagnóstico por imagem , Corantes Fluorescentes/química
13.
Front Pharmacol ; 12: 670207, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995101

RESUMO

Ovarian cancer has the highest death rate in gynecologic tumors and the main therapy for patients with advanced is chemotherapy based on cisplatin. Additionally, hyperthermic intraperitoneal has been used in clinic to obtain better efficacy for patients. Hence, combined photothermal therapy with platinum drugs in a new delivery system might bring new hope for ovarian cancer. A reduction sensitive polymer encapsulating a Pt (IV) prodrug and a near infrared II (NIR II) photothermal agent IR1048 to form nanoparticles were reported to enhance the efficacy of ovarian cancer treatment. At the same time, endoplasmic reticulum stress indicates an imbalance in proteostasis which probably caused by extrinsic stress such as chemotherapy and the temperature changed. The efficacy of nanoparticles containing Pt (IV) and IR1048 under NIR II light might be caused via increased DNA damage and endoplasmic reticulum (ER) stress.

14.
Adv Mater ; 33(20): e2100599, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33834553

RESUMO

A systematic combination strategy is proposed for overcoming cisplatin resistance using near-infrared (NIR)-light-triggered hyperthermia. A new photothermal polymer DAP-F is complexed with a reduction-sensitive amphiphilic polymer P1 to form F-NPs with photothermal effect. Subsequently, to build the final nanosystem F-Pt-NPs, F-NPs are combined with Pt-NPs, which are obtained by encapsulating a Pt(IV) prodrug with P1. Mild hyperthermia (43 °C), generated from F-Pt-NPs induced by an 808 nm NIR laser, have various effects such as: i) enhancing the cellular membrane permeability to promote the uptake of drugs; ii) activating cisplatin by accelerating the glutathione consumption; iii) increasing the Pt-DNA adducts formation and possibly the formation of a portion of irreparable Pt-DNA interstrand crosslinks, thereby inhibiting the repair of DNA. In vitro, it is found that even on cisplatin-resistant A549DDP cells, the IC50 of F-Pt-NPs (43 °C) is only 7.0 × 10-6 m Pt mL-1 . In vivo, on a patient-derived xenograft model of multidrug resistant lung cancer, the efficacy of the F-Pt-NPs (43 °C) treatment group shows a tumor inhibition rate of 94%. Taken together, here, an important perspective of resolving cascade drug resistance with the assistance of mild hyperthermia triggered by NIR light is presented, which can be of great significance for clinic translation.


Assuntos
Antineoplásicos , Sistemas de Liberação de Medicamentos , Animais , Cisplatino , Hipertermia , Pró-Fármacos
15.
J Mater Chem B ; 9(26): 5173-5194, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34116565

RESUMO

Platinum drugs are commonly used in cancer therapy, but their therapeutic outcomes have been significantly compromised by the drug resistance of cancer cells. To this end, intensive efforts have been made to develop nanoparticle-based drug delivery systems for platinum drugs, due to their multifunctionality in delivering drugs, in modulating the tumor microenvironment, and in integrating additional genes, proteins, and small molecules to overcome chemoresistance in cancers. To facilitate the clinical application of these promising nanoparticle-based platinum drug delivery systems, this paper summarizes the common mechanisms for chemoresistance towards platinum drugs, the advantages of nanoparticles in drug delivery, and recent strategies of nanoparticle-based platinum drug delivery. Furthermore, we discuss how to design delivery platforms more effectively to overcome chemoresistance in cancers, thereby improving the efficacy of platinum-based chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Antineoplásicos/química , Humanos , Compostos Organoplatínicos/química
16.
ACS Nano ; 15(3): 5428-5438, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33689300

RESUMO

NIR-II (1000-1700 nm) fluorescence imaging is continually attracting strong research interest. However, current NIR-II imaging materials are limited to small molecules with fast blood clearance and inorganic nanomaterials and organic conjugated polymers of poor biodegradability and low biocompatibility. Here, we report a highly biodegradable polyester carrying tandem NIR-II fluorophores as a promising alternative. The polymer encapsulated a platinum intercalator (56MESS, (5,6-dimethyl-1,10-phenanthroline) (1S,2S-diaminocyclohexane) platinum(II)) and was conjugated with both a cell-targeting RGD peptide and a caspase-3 cleavable peptide probe to form nanoparticles for simultaneous NIR-II and apoptosis imaging. In vitro, the nanoparticles were approximately 4-1000- and 1.5-10-fold more potent than cisplatin and 56MESS, respectively. Moreover, in vivo, they significantly inhibited tumor growth on a multidrug-resistant patient-derived mouse model (PDXMDR). Finally, through label-free laser desorption-ionization mass spectrometry imaging (MALDI-MSI), in situ 56MESS release in the deeper tumors was observed. This work highlighted the use of biodegradable NIR-II polymers for monitoring drugs in vivo and therapeutic effect feedback in real-time.


Assuntos
Nanopartículas , Preparações Farmacêuticas , Animais , Linhagem Celular Tumoral , Retroalimentação , Humanos , Camundongos , Polímeros , Resultado do Tratamento
17.
Nat Biomed Eng ; 5(9): 1048-1058, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34045730

RESUMO

In patients with glioblastoma, resistance to the chemotherapeutic temozolomide (TMZ) limits any survival benefits conferred by the drug. Here we show that the convection-enhanced delivery of nanoparticles containing disulfide bonds (which are cleaved in the reductive environment of the tumour) and encapsulating an oxaliplatin prodrug and a cationic DNA intercalator inhibit the growth of TMZ-resistant cells from patient-derived xenografts, and hinder the progression of TMZ-resistant human glioblastoma tumours in mice without causing any detectable toxicity. Genome-wide RNA profiling and metabolomic analyses of a glioma cell line treated with the cationic intercalator or with TMZ showed substantial differences in the signalling and metabolic pathways altered by each drug. Our findings suggest that the combination of anticancer drugs with distinct mechanisms of action with selective drug release and convection-enhanced delivery may represent a translational strategy for the treatment of TMZ-resistant gliomas.


Assuntos
Neoplasias Encefálicas , Glioma , Nanopartículas , Animais , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Convecção , DNA , Glioma/tratamento farmacológico , Humanos , Substâncias Intercalantes , Camundongos , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mater Sci Eng C Mater Biol Appl ; 111: 110855, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32279770

RESUMO

Bacterial adhesion and colonization on material surfaces have attracted great attention due to their potential threat to human health. Combining bactericidal and antifouling functions has been confirmed as an optimal strategy to prevent microbial infection. In this work, biodegradable electrospun polyvinyl alcohol (PVA) nanofibers were chosen due to its high specific area and abundant reactive hydroxyl groups. A quaternary ammonium salt (IQAS) and zwitterionic sulfopropylbetaine (ISB), both containing isocyanate (NCO) groups, were chemically bonded to the PVA nanofiber surface via a coupling reaction between the OH groups of the PVA nanofibers and the NCO groups of IQAS or ISB. The results indicated that the antimicrobial rates of PVA nanofibers modified by IQAS (0.5%) reached 99.9% against both gram-positive Staphylococcus aureus (S. aureus, ATCC 6538) and gram-negative Escherichia coli (E. coli, ATCC 25922). Additionally, the live/dead staining and cytotoxicity test indicated that the dual functional IQAS/ISB/PVA nanofibers exhibited excellent bactericidal and antifouling activities with low cytotoxicity. This work may provide practical guidelines to fabricate bactericidal and antifouling materials for healthcare applications, including but not limited to wound dressings, textile, food packaging and air filtration.


Assuntos
Antibacterianos/farmacologia , Betaína/farmacologia , Incrustação Biológica , Nanofibras/química , Álcool de Polivinil/farmacologia , Compostos de Amônio Quaternário/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Células NIH 3T3 , Álcool de Polivinil/química , Espectroscopia de Prótons por Ressonância Magnética , Resistência à Tração
19.
ACS Nano ; 14(12): 16984-16996, 2020 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-33283501

RESUMO

Excessive oxidative stress in cancer cells can induce cancer cell death. Anticancer activity and drug resistance of chemotherapy are closely related to the redox state of tumor cells. Herein, five lipophilic Pt(IV) prodrugs were synthesized on the basis of the most widely used anticancer drug cisplatin, whose anticancer efficacy and drug resistance are closely related to the intracellular redox state. Subsequently, a series of cisplatin-sensitive and drug-resistant cell lines as well as three patient-derived primary ovarian cancer cells have been selected to screen those prodrugs. To verify if the disruption of redox balance can be combined with these Pt(IV) prodrugs, we then synthesized a polymer with a diselenium bond in the main chain for encapsulating the most effective prodrug to form nanoparticles (NP(Se)s). NP(Se)s can efficiently break the redox balance via simultaneously depleting GSH and augmenting ROS, thereby achieving a synergistic effect with cisplatin. In addition, genome-wide analysis via RNA-seq was employed to provide a comprehensive understanding of the changes in transcriptome and the alterations in redox-related pathways in cells treated with NP(Se)s and cisplatin. Thereafter, patient-derived xenograft models of hepatic carcinoma (PDXHCC) and multidrug-resistant lung cancer (PDXMDR) were established to evaluate the therapeutic effect of NP(Se)s, and a significant antitumor effect was achieved on both models with NP(Se)s. Overall, this study provides a promising strategy to break the redox balance for maximizing the efficacy of platinum-based cancer therapy.

20.
ACS Nano ; 14(11): 14831-14845, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33084319

RESUMO

DNA alkylating agents generally kill tumor cells by covalently binding with DNA to form interstrand or intrastrand cross-links. However, in the case of cisplatin, only a few DNA adducts (<1%) are highly toxic irreparable interstrand cross-links. Furthermore, cisplatin is rapidly detoxified by high levels of intracellular thiols such as glutathione (GSH). Since the discovery of its mechanism of action, people have been looking for ways to directly and efficiently remove intracellular GSH and increase interstrand cross-links to improve drug efficacy and overcome resistance, but there has been little breakthrough. Herein, we hypothesized that the anticancer efficiency of cisplatin can be enhanced through iodo-thiol click chemistry mediated GSH depletion and increased formation of DNA interstrand cross-links via mild hyperthermia triggered by near-infrared (NIR) light. This was achieved by preparing an amphiphilic polymer with platinum(IV) (Pt(IV)) prodrugs and pendant iodine atoms (iodides). The polymer was further used to encapsulate IR780 and assembled into Pt-I-IR780 nanoparticles. Induction of mild hyperthermia (43 °C) at the tumor site by NIR light irradiation had three effects: (1) it accelerated the GSH-mediated reduction of Pt(IV) in the polymer main chain to platinum(II) (Pt(II)); (2) it boosted the iodo-thiol substitution click reaction between GSH and iodide, thereby attenuating the GSH-mediated detoxification of cisplatin; (3) it increased the proportion of highly toxic and irreparable Pt-DNA interstrand cross-links. Therefore, we find that mild hyperthermia induced via NIR irradiation can enhance the killing of cancer cells and reduce the tumor burden, thus delivering efficient chemotherapy.


Assuntos
Antineoplásicos , Cisplatino , Reagentes de Ligações Cruzadas , Adutos de DNA , Glutationa , Hipertermia Induzida , Antineoplásicos/farmacologia , Cisplatino/farmacologia , DNA/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA