Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nano Lett ; 23(10): 4216-4225, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37155369

RESUMO

Adjuvant whole-breast radiotherapy is essential for breast cancer patients who adopted breast-conserving surgery (BCS) to reduce the risk of local recurrences, which however suffer from large-area and highly destructive ionizing radiation-induced adverse events. To tackle this issue, an afterglow/photothermal bifunctional polymeric nanoparticle (APPN) is developed that utilizes nonionizing light for precise afterglow imaging-guided post-BCS adjuvant second near-infrared (NIR-II) photothermal therapy. APPN consists of a tumor cell targeting afterglow agent, which is doped with a NIR dye as an afterglow initiator and a NIR-II light-absorbing semiconducting polymer as a photothermal transducer. Such a design realizes precise afterglow imaging-guided NIR-II photothermal ablation of minimal residual breast tumor foci after BCS, thus achieving complete inhibition of local recurrences. Moreover, APPN enables early diagnosis and treatment of local recurrence after BCS. This study thus provides a nonionizing modality for precision post-BCS adjuvant therapy and early recurrence theranostic.


Assuntos
Nanopartículas , Medicina de Precisão , Humanos , Fototerapia , Polímeros , Recidiva , Linhagem Celular Tumoral
2.
Nanoscale ; 15(13): 6252-6262, 2023 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-36908261

RESUMO

The need for adjuvant therapy to inhibit local recurrence after breast-conserving surgery with minimal side effects is great. Adjuvant photothermal therapy (aPTT) has the potential to replace radiotherapy and eliminates its inherent damage to healthy tissues. Herein, we functionalized semiconducting polymer nanoparticles (SPNs) with cRGD-peptide and silicon 2,3-naphthalocyanine bis(trihexylsilyloxide) (NIR775) to target breast cancer and perform aPTT under an ultra-low laser power (0.2 W cm-2) after breast-conserving surgery (BCS). The synthesized RGD-SPNNIR775 showed an excellent photothermal conversion efficiency and biocompatibility and was demonstrated to accumulate in tumors specifically. The BCS could be performed with confidence under the guidance of preoperative and postoperative fluorescence imaging. Notably, the aPTT completely inhibited the local recurrence after the BCS without compromising the cosmetic effect of the BCS. These results indicate the prospect of RGD-SPNNIR775 as a theranostic nanoplatform for efficient aPTT using an ultra-low laser power to control recurrence after BCS.


Assuntos
Neoplasias da Mama , Nanopartículas , Humanos , Feminino , Terapia Fototérmica , Polímeros/farmacologia , Mastectomia Segmentar/métodos , Neoplasias da Mama/patologia , Adjuvantes Imunológicos , Nanopartículas/uso terapêutico , Lasers , Recidiva , Oligopeptídeos/farmacologia
3.
ACS Appl Mater Interfaces ; 14(25): 28570-28580, 2022 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-35726862

RESUMO

Due to the strong and tunable photothermal effect, metallic nanoparticles are of enormous interest in light-activated biomedical applications, such as photoacoustic imaging (PAI) and photothermal therapy (PTT). However, the photothermal conversion efficiency (PCE) of existing metallic photothermal agents is still unsatisfactory. Herein, we develop an efficient photothermal theranostic agent based on a gold nanostar@polyaniline core-shell nanocomposite with high PCE for PAI-guided PTT at a low dosage. After optimizing the relative composition of polyaniline (PANI) and gold nanostars (AuNSs), this nanocomposite eventually empowers an outstanding PCE of up to 78.6%, which is much better than AuNSs or PANI alone and most of the existing photothermal theranostic agents. Besides, the nanocomposite can act as a targeted probe for tumors by hyaluronic acid (HA) modification without compromising the photothermal performance. The obtained nanoprobes named AuNSPHs exhibit promising biocompatibility and great performance of PAI-guided PTT to treat triple-negative breast cancer both in vitro and in vivo. More importantly, a single injection of AuNSPHs significantly suppresses tumor growth with a low dosage of Au (0.095 mg/kg), which is attributed to the high PCE of AuNSPHs. Taking advantage of the exhilarating photothermal conversion ability, this theranostic agent can safely potentiate the antitumor therapeutic efficacy of laser-induced ablation and holds great potential for future medical applications.


Assuntos
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Compostos de Anilina , Ouro/farmacologia , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Técnicas Fotoacústicas/métodos , Fototerapia , Medicina de Precisão , Nanomedicina Teranóstica/métodos
4.
J Nanobiotechnology ; 19(1): 365, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789274

RESUMO

BACKGROUND: Tumor phototherapy especially photodynamic therapy (PDT) or photothermal therapy (PTT), has been considered as an attractive strategy to elicit significant immunogenic cell death (ICD) at an optimal tumor retention of PDT/PTT agents. Heptamethine cyanine dye (IR-780), a promising PDT/PTT agent, which can be used for near-infrared (NIR) fluorescence/photoacoustic (PA) imaging guided tumor phototherapy, however, the strong hydrophobicity, short circulation time, and potential toxicity in vivo hinder its biomedical applications. To address this challenge, we developed mesoporous polydopamine nanoparticles (MPDA) with excellent biocompatibility, PTT efficacy, and PA imaging ability, facilitating an efficient loading and protection of hydrophobic IR-780. RESULTS: The IR-780 loaded MPDA (IR-780@MPDA) exhibited high loading capacity of IR-780 (49.7 wt%), good physiological solubility and stability, and reduced toxicity. In vivo NIR fluorescence and PA imaging revealed high tumor accumulation of IR-780@MPDA. Furthermore, the combined PDT/PTT of IR-780@MPDA could induce ICD, triggered immunotherapeutic response to breast tumor by the activation of cytotoxic T cells, resulting in significant suppression of tumor growth in vivo. CONCLUSION: This study demonstrated that the as-developed compact and biocompatible platform could induce combined PDT/PTT and accelerate immune activation via excellent tumor accumulation ability, offering multimodal tumor theranostics with negligible systemic toxicity.


Assuntos
Antineoplásicos , Carbocianinas , Corantes Fluorescentes , Indóis/química , Nanopartículas/química , Polímeros/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Carbocianinas/química , Carbocianinas/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Neoplasias Mamárias Animais , Camundongos , Fototerapia , Nanomedicina Teranóstica , Distribuição Tecidual
5.
Signal Transduct Target Ther ; 5(1): 87, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32532960

RESUMO

Sorafenib is a multikinase inhibitor capable of facilitating apoptosis, mitigating angiogenesis and suppressing tumor cell proliferation. In late-stage hepatocellular carcinoma (HCC), sorafenib is currently an effective first-line therapy. Unfortunately, the development of drug resistance to sorafenib is becoming increasingly common. This study aims to identify factors contributing to resistance and ways to mitigate resistance. Recent studies have shown that epigenetics, transport processes, regulated cell death, and the tumor microenvironment are involved in the development of sorafenib resistance in HCC and subsequent HCC progression. This study summarizes discoveries achieved recently in terms of the principles of sorafenib resistance and outlines approaches suitable for improving therapeutic outcomes for HCC patients.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Hepáticas/tratamento farmacológico , Sorafenibe/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Sorafenibe/efeitos adversos , Microambiente Tumoral/efeitos dos fármacos
6.
Biomaterials ; 232: 119677, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31865193

RESUMO

Conventional radiotherapy has a pivotal role in the treatment of glioblastoma; nevertheless, its clinical utility has been limited by radiation resistance. There is emerging evidence that upregulated heat shock protein A5 (HSPA5) in cancer cells maintains or restores the homeostasis of a cellular microenvironment and results in cancer resistance in various treatments. Therefore, we describe a bioresponsive nanoplatform that can deliver a HSPA5 inhibitor (pifithrin-µ, PES) and radiosensitizer (gold nanosphere, AuNS), to expand the synergistic photothermal therapy and radiotherapy, as well as to monitor the progression of cancer therapy using computer tomography/magnetic resonance imaging. The nanoplatform (PES-Au@PDA, 63.3 ± 3.1 nm) comprises AuNS coated with the photothermal conversion agent polydopamine (PDA) for enhanced radiotherapy and photothermal therapy, as well as PES (loading efficiency of PES approximately 40%), a small molecular inhibitor against HSPA5 to amplify the pro-apoptotic unfolded protein response (UPR). The reported nanoplatform enables hyperthermia-responsive release of PES. Results from in vitro and in vivo studies demonstrate that PES-Au@PDA can specially activate pro-apoptotic UPR cascades, leading to remarkably improved radiotherapy and photothermal therapy efficiencies. Considered together, a versatile theranostic nanosystem is reported for promoting the synergistic radiophotothermal therapy by selectively activating pro-apoptotic UPR cascade pathways.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Hipertermia Induzida , Nanopartículas Metálicas , Neoplasias Encefálicas/tratamento farmacológico , Chaperona BiP do Retículo Endoplasmático , Glioblastoma/tratamento farmacológico , Ouro , Humanos , Fototerapia , Sulfonamidas , Microambiente Tumoral , Resposta a Proteínas não Dobradas
7.
ACS Appl Mater Interfaces ; 11(50): 46626-46636, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31751121

RESUMO

Programmed cell death ligand 1 (PD-L1) blockade has achieved great success in cancer immunotherapy; however, the response of triple-negative breast cancer (TNBC) to PD-L1 antibodies is limited. To address this challenge, we use the bromodomain and extra-terminal inhibitor JQ1 to down-regulate the expression of PD-L1 and thus elicit the immune response to TNBC instead of using antibodies to block PD-L1. JQ1 also inhibits the growth of TNBC as a targeted therapeutic agent by inhibiting the BRD4-c-MYC axis. The polydopamine nanoparticles (PDMNs) are introduced as a biodegradable and adaptable platform to load JQ1 and induce photothermal therapy (PTT) as another synergistic therapeutic modality. Because the JQ1-loaded PDMNs (PDMN-JQ1) are self-degradable and release JQ1 continuously, this synergistic treatment can lead to remarkable activation of cytotoxic T lymphocytes and induce a strong immune-memory effect to protect mice from tumor re-challenge. Taken together, our study demonstrates a compact and simple nanoplatform for triple therapy, including targeted therapy, PTT, and immunotherapy, for TNBC treatment.


Assuntos
Antígeno B7-H1/genética , Nanopartículas/química , Fototerapia , Proteínas Proto-Oncogênicas c-myc/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Anticorpos/genética , Apoptose/efeitos dos fármacos , Azepinas/química , Azepinas/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Xenoenxertos , Humanos , Indóis/química , Indóis/farmacologia , Terapia com Luz de Baixa Intensidade , Camundongos , Polímeros/química , Polímeros/farmacologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Triazóis/química , Triazóis/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
8.
ACS Nano ; 13(2): 2544-2557, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30730695

RESUMO

Combination therapy, especially photodynamic/photothermal therapy (PDT/PTT), has shown promising applications in cancer therapy. However, sequential irradiation by two different laser sources and even the utilization of single high-power laser to induce either combined PDT/PTT or individual PTT will be subjected to prolonged treatment time, complicated treatment process, and potential skin burns. Thus, low power single laser activatable combined PDT/PTT is still a formidable challenge. Herein, we propose an effective strategy to achieve synergistic cancer phototherapy under low power single laser irradiation for short duration. By taking advantage of dual plasmonic PTT nanoagents (AuNRs/MoS2), a significant increase in temperature up to 60 °C with an overall photothermal conversion efficiency (PCE) of 68.8% was achieved within 5 min under very low power (0.2 W/cm2) NIR laser irradiation. The enhanced PCE and PTT performance is attributed to the synergistic plasmonic PTT effect (PPTT) of dual plasmonic nanoagents, promoting simultaneous release (85%) of electrostatically bonded indocyanine green (ICG) to induce PDT effects, offering simultaneous PDT/synergistic PPTT. Both in vitro and in vivo investigations reveal complete cell/tumor eradication, implying that simultaneous PDT/synergistic PPTT effects induced by AuNRs/MoS2-ICG are much superior over individual PDT or synergistic PPTT. Notably, synergistic PPTT induced by dual plasmonic nanoagents also demonstrates higher in vivo antitumor efficacy than either individual PDT or PTT agents. Taken together, under single laser activation with low power density, the proposed strategy of simultaneous PDT/synergistic PPTT effectively reduces the treatment time, achieves high therapeutic index, and offers safe treatment option, which may serve as a platform to develop safer and clinically translatable approaches for accelerating cancer therapeutics.


Assuntos
Lasers , Nanopartículas/química , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Proliferação de Células/efeitos dos fármacos , Dissulfetos/química , Dissulfetos/farmacologia , Relação Dose-Resposta a Droga , Ouro/química , Ouro/farmacologia , Células HeLa , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Molibdênio/química , Molibdênio/farmacologia , Imagem Óptica , Fármacos Fotossensibilizantes/química
9.
ACS Appl Mater Interfaces ; 10(20): 16992-17003, 2018 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-29722264

RESUMO

Non-small cell lung cancer (NSCLC) is difficult to cure because of the high recurrence rate and the side effects of current treatments. It is urgent to develop a new treatment that is safer and more effective than current treatments against NSCLC. Herein, we constructed anti-epidermal growth factor receptor (EGFR) peptide-conjugated PEGylated triangular gold nanoplates (TGN-PEG-P75) as a targeting photothermal therapy (PTT) agent to treat NSCLC under the guidance of computed tomography (CT) and photoacoustic (PA) imaging. The surface of TGNs is successfully conjugated with a novel peptide P75 that has the specific affinity to epidermal growth factor receptor (EGFR). It is found that the EGFR is overexpressed in NSCLC cells. The TGN-PEG-P75 has uniform edge length (77.9 ± 7.0 nm) and neutrally charged surface. The cell uptake experiments demonstrate remarkable affinity of the TGN-PEG-P75 to high EGFR expression cells than low EGFR expression cells (5.1-fold). Thanks to the strong near-infrared absorbance, high photothermal conversion efficiency, and the increased accumulation in tumor cells via the interaction of P75 and EGFR, TGN-PEG-P75 exhibits 3.8-fold superior therapeutic efficacy on HCC827 cells than TGN-PEG. The in vivo CT/PA dual-modal imaging of the TGN-PEG-P75 is helpful in selecting the optimal treatment time and providing real-time visual guidance of PTT. Furthermore, treatments on HCC827 tumor-bearing mouse model demonstrate that the growth of NSCLC cells can be effectively inhibited by the TGN-PEG-P75 through PTT, indicating the great promise of the nanoplatform for treating NSCLC in vivo.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Receptores ErbB , Ouro , Camundongos , Peptídeos , Técnicas Fotoacústicas , Fototerapia
10.
J Mater Chem B ; 6(23): 3865-3875, 2018 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-32254314

RESUMO

A variety of nanocarriers have been designed to deliver photosensitizers (PSs) and promote the clinical applications of photodynamic therapy (PDT). However, most of them suffer from insufficient loading capability, premature leakage, and/or unstable therapeutic efficacy. Herein, we constructed a novel nanocomposite (TGP@MOS) with a benzene-bridged mesoporous organosilica shell and a triangular gold nanoprism core. The TGP@MOS could load model PS molecules, zinc phthalocyanine (ZnPc), with high loading capacity (11.8 wt%) and minimal premature leakage (only 2.6% after incubation in PBS with 10% FBS for 60 h) viaπ-π stacking interactions and hydrophobic interactions. We demonstrated that the obtained TGP@MOS-ZnPc could realize timely coordinated photodynamic/photothermal therapy upon single irradiation, and thus stabilize and maximize the therapeutic efficacy of phototherapy both in vitro and in vivo. Other advantages of TGP@MOS-ZnPc include excellent water solubility, stability, hemocompatibility and biocompatibility.

11.
RSC Adv ; 8(56): 32200-32210, 2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-35547489

RESUMO

Photodynamic therapy (PDT) has attracted much attention as a strategy for tumor therapy. However, the insolubility and poor tumor-targeting ability of most photosensitizers (PSs) hinder PDT from further development. Therefore, it is necessary to explore new carriers with good water solubility and biocompatibility to deliver PSs to tumors. Melanin nanoparticles are novel biomimetic nanocarriers with excellent biocompatibility, loading capacity, photothermal therapy (PTT) and magnetic resonance (MR)/photoacoustic (PA) imaging properties. Here we designed polydopamine melanin nanoparticles (PDMNs) as a delivery platform for the photosensitizer Chlorin e6 (PDMN-Ce6) and realized its application as a theranostic agent for tumor therapy. The PDMN-Ce6 exhibited excellent biocompatibility, good water solubility and high loading capability (35.2 wt%) for Ce6. Compared with the free Ce6, PDMN-Ce6 showed higher cellular internalization and superior synergistic phototherapy effects in an in vitro study. An in vivo study indicated that the accumulation of PDMN-Ce6 at tumor sites was 2.8-fold higher than that of free Ce6 at 24 h post-injection, which was beneficial for MR/PA imaging. Moreover, the synergetic therapy significantly inhibited tumor growth, causing tumor necrosis and tumor angiogenesis suppression. These results suggest that our biomimetic and biocompatible platform could improve the delivery of Ce6 to tumors and realize multimodal imaging-guided tumor synergetic phototherapy.

12.
ACS Nano ; 12(1): 662-670, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29271636

RESUMO

Adjuvant treatments following breast-conserving surgery (BCS) are essential to reduce the risk of local recurrences in patients with breast cancer. However, current adjuvant treatments are based on ionizing radiation, which brings radiation-induced damage and amplifies the risk of death. Here we explore the feasibility of using non-ionizing light to induce photothermal therapy as an adjuvant treatment to BCS. In an orthotopic breast cancer mice model, we demonstrate that adjuvant photothermal therapy (aPTT) decreases the incidence of local recurrences after BCS with no expense of cosmetic outcome. In comparison with conventional photothermal therapy, the technique used in aPTT provides more uniformly distributed light energy and less risk of skin burns and local recurrences. Overall, this work represents a departure from the traditional concept of using PTT as an alternative to surgery and reveals the potential of using PTT as an alternative to adjuvant radiation therapy, which is valuable especially for patients susceptible to radiation damage.


Assuntos
Neoplasias da Mama/cirurgia , Neoplasias da Mama/terapia , Recidiva Local de Neoplasia/prevenção & controle , Animais , Linhagem Celular Tumoral , Terapia Combinada/métodos , Feminino , Humanos , Hipertermia Induzida/métodos , Mastectomia Segmentar/métodos , Camundongos Nus , Fototerapia/métodos , Resultado do Tratamento
13.
Nanoscale ; 9(4): 1466-1474, 2017 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-28066849

RESUMO

To date, clinicians still lack an effective strategy to treat triple negative breast cancer (TNBC). In this work, we design for the first time a gold nanorod embedded large-pore mesoporous organosilica (GNR@LPMO) nanoplatform for gene and photothermal cooperative therapy of TNBC. The synthesized GNR@LPMOs possess a uniform size (175 nm), high surface area (631 m2 g-1), large pore size, excellent photothermal efficiency, and good biocompatibility. Thanks to the large-pore mesoporous organosilica layer, the GNR@LPMO nanoplatforms display much higher loading capacity of siRNA compared with traditional liposome and bare gold nanorods. Thus, functional siRNA can be efficiently delivered into TNBC cells by GNR@LPMOs, causing much higher cell apoptosis through knocking down the PLK1 proteins. By combining the effective gene delivery and photothermal abilities, the GNR@LPMO nanoplatforms are further used for gene and photothermal cooperative therapy of TNBC, which induce a 15 fold higher mice tumor inhibition rate than sole therapy modality, indicating the potential clinical use of this novel nanoplatform in treating TNBC.


Assuntos
Terapia Genética , Ouro , Nanosferas , Fototerapia , Neoplasias de Mama Triplo Negativas/terapia , Animais , Apoptose , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Temperatura Alta , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanotubos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Quinase 1 Polo-Like
14.
ACS Nano ; 10(9): 8578-90, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27576159

RESUMO

Selectively enhance the therapeutic efficacy to malignancy is one of the most important issues for photothermal therapy (PTT). However, most solid tumors, such as triple negative breast cancer (TNBC), do not have identifiable surface markers to distinguish themselves from normal cells, thus it is challenging to selectively identify and eliminate those malignances by PTT. In this report, we hypothesized that, by targeting CD44 (one TNBC-overexpressed surface molecule) and depleting heat shock protein 72 (HSP72, one malignancy-specific-overexpressed thermotolerance-related chaperone) subsequently, the TNBC could be selectively sensitized to PTT and improve the accuracy of treatment. To this end, a rationally designed nanosystem gold nanostar (GNS)/siRNA against HSP72 (siHSP72)/hyaluronic acid (HA) was successfully constructed using a layer-by-layer method. Hydrodynamic diameter and zeta potential analysis demonstrated the formation of GNS/siHSP72/HA having a particle size of 73.2 ± 3.8 nm and a negative surface charge of -18.3 ± 1.6 mV. The CD44-targeting ability of GNS/siHSP72/HA was confirmed by the flow cytometer, confocal microscopic imaging, and competitive binding analysis. The HSP72 silencing efficacy of GNS/siHSP72/HA was ∼95% in complete culture medium. By targeting CD44 and depleting HSP72 sequentially, GNS/siHSP72/HA could selectively sensitize TNBC cells to hyperthermia and enhance the therapeutic efficacy to TNBC with minimal side effect both in vitro and in vivo. Other advantages of GNS/siHSP72/HA included easy synthesis, robust siRNA loading capacity, endosome/lysosome escaping ability, high photothermal conversion efficacy and superior hemo- and biocompatibility.


Assuntos
Proteínas de Choque Térmico HSP72/metabolismo , Receptores de Hialuronatos/metabolismo , Nanopartículas Metálicas , Fototerapia , Transporte Proteico , Linhagem Celular Tumoral , Ouro , Humanos , Hipertermia Induzida , Neoplasias/terapia
15.
ACS Appl Mater Interfaces ; 8(28): 17927-35, 2016 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-27356586

RESUMO

Mesenchymal stem cells (MSCs) have attracted increasing attention as vehicles for cancer treatment. Herein, MSC-based synergistic oncotherapy strategy is presented for the first time. To achieve this goal, yolk-shell structured gold nanorod embedded hollow periodic mesoporous organosilica nanospheres (GNR@HPMOs) with high paclitaxel (PTX) loading capability and excellent photothermal transfer ability upon near-infrared (NIR) light irradiation are first prepared. Cytotoxicity and migration assays show that the viability and tumor-homing capability of MSCs are well-retained after internalization of high content of PTX loaded GNR@HPMOs (denoted as GNR@HPMOs-PTX). In vitro experiments show the GNR@HPMOs-PTX loaded MSCs (GNR@HPMOs-PTX@MSCs) possess synergistic chemo-photothermal killing effects for breast cancer cells. Also, photoacoustic imaging shows that the MSCs can improve dispersion and distribution in tumor tissue for GNR@HPMOs-PTX after intratumoral injection. In vivo experiments in breast cancer model of nude mice further demonstrate that the GNR@HPMOs-PTX@MSCs significantly inhibit tumor growth, suggesting their great potential for synergistic therapy of cancer.


Assuntos
Neoplasias da Mama/terapia , Sistemas de Liberação de Medicamentos/métodos , Hipertermia Induzida/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Paclitaxel/administração & dosagem , Fototerapia/métodos , Animais , Neoplasias da Mama/tratamento farmacológico , Feminino , Ouro/química , Humanos , Células MCF-7 , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanoconchas/administração & dosagem , Nanoconchas/química , Nanosferas/química , Nanotubos/química , Compostos de Organossilício/química , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Nanoscale ; 7(14): 6304-10, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25782595

RESUMO

We have developed a novel aptamer-targeting photoresponsive drug delivery system by non-covalent assembly of a Cy5.5-AS1411 aptamer conjugate on the surface of graphene oxide wrapped doxorubicin (Dox)-loaded mesoporous silica nanoparticles (MSN-Dox@GO-Apt) for light-mediated drug release and aptamer-targeted cancer therapy. The two "off-on" switches of the MSN-Dox@GO-Apt were controlled by aptamer targeting and light triggering, respectively. The Cy5.5-AS1411 ligand provides MSN-Dox@GO-Apt with nucleolin specific targeting and real-time indicator abilities by "off-on" Cy5.5 fluorescence recovery. The GO acts as a gatekeeper to prevent the loaded Dox from leaking in the absence of laser irradiation, and to control the Dox release in response to laser irradiation. When the GO wrapping falls off upon laser irradiation, the "off-on" photoresponsive drug delivery system is activated, thus inducing chemotherapy. Interestingly, with an increase in laser power, the synergism of chemotherapy and photothermal therapy in a single MSN-Dox@GO-Apt platform led to much more effective cancer cell killing than monotherapies, providing a new approach for treatment against cancer.


Assuntos
Aptâmeros de Nucleotídeos/química , Doxorrubicina , Grafite/química , Nanopartículas/química , Neoplasias/terapia , Fototerapia , Dióxido de Silício/química , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Luz , Células MCF-7 , Neoplasias/metabolismo , Neoplasias/patologia , Porosidade
17.
Small ; 10(8): 1585-93, 1441, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24150920

RESUMO

Photoacoustic (PA) imaging promises deeper tissue penetration while maintaining rich optical contrast as compared to other high resolution optical imaging techniques. In this report, a near-infrared pulse laser serves as the excitation source, and 128 ultrasonic transducers are spirally distributed on a hemispherical surface to receive PA signals for three-dimensional (3D) image reconstruction. With these attributes, the unique modality produces an isotropic and homogeneous spatial resolution (∼200 µm) with penetration depth of centimeters. Cyclic Arg-Gly-Asp (RGD) peptides conjugated plasmonic gold nanostars (RGD-GNS) are designed to specifically target over-expressed integrin α(v)ß3 on tumor neovasculature, enabling highly sensitive angiography and photothermal therapy (PTT). After the administration of RGD-GNS, tumor angiogenesis is clearly imaged with enhanced contrast, and the growth of tumor is effectively inhibited by PTT after laser irradiation. This study suggest that the PA angiography with plasmonic RGD-GNS can be applied as a triple functional platform for tumor diagnosis, PTT, and treatment monitoring. This PA technique offers deeper imaging depth with homogeneous resolution over existing optical imaging techniques for early diagnosis of tumor angiogenesis as well as on-the-spot nanotherapeutic evaluation.


Assuntos
Angiografia/métodos , Nanopartículas Metálicas , Neovascularização Patológica/diagnóstico , Técnicas Fotoacústicas/métodos , Animais , Linhagem Celular Tumoral , Meios de Contraste , Glioma/irrigação sanguínea , Glioma/diagnóstico , Glioma/terapia , Ouro , Humanos , Hipertermia Induzida , Imageamento Tridimensional , Integrina alfaVbeta3/metabolismo , Nanopartículas Metálicas/ultraestrutura , Camundongos , Camundongos Nus , Oligopeptídeos , Imagens de Fantasmas
18.
Angew Chem Int Ed Engl ; 52(52): 13958-13964, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24318645

RESUMO

The hierarchical assembly of gold nanoparticles (GNPs) allows the localized surface plasmon resonance peaks to be engineered to the near-infrared (NIR) region for enhanced photothermal therapy (PTT). Herein we report a novel theranostic platform based on biodegradable plasmonic gold nanovesicles for photoacoustic (PA) imaging and PTT. The disulfide bond at the terminus of a PEG-b-PCL block-copolymer graft enables dense packing of GNPs during the assembly process and induces ultrastrong plasmonic coupling between adjacent GNPs. The strong NIR absorption induced by plasmon coupling and very high photothermal conversion efficiency (η=37%) enable simultaneous thermal/PA imaging and enhanced PTT efficacy with improved clearance of the dissociated particles after the completion of PTT. The assembly of various nanocrystals with tailored optical, magnetic, and electronic properties into vesicle architectures opens new possibilities for the construction of multifunctional biodegradable platforms for biomedical applications.


Assuntos
Diagnóstico por Imagem/métodos , Ouro/química , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Humanos , Nanopartículas , Resultado do Tratamento
19.
PLoS One ; 7(12): e53449, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285296

RESUMO

BACKGROUND: Malignant pleural effusion (MPE) is a common complication of lung cancer. One widely used treatment for MPE is Endostar, a recombined humanized endostatin based treatment. However, the mechanism of this treatment is still unclear. The aim of this study was to investigate the effects of Endostar in mice with MPE. METHODS AND MATERIALS: Lewis lung carcinoma (LLC) cell line expressing enhanced green fluorescent protein (EGFP) was injected into pleural cavity to establish MPE mice model. Mice were randomly divided into four groups. High dose of Endostar (30 mg/kg), low dose of Endostar (8 mg/kg), normal saline, or Bevacizumab (5 mg/kg) was respectively injected into pleural cavity three times with 3-day interval in each group. Transverse computed tomography (CT) was performed to observe pleural fluid formation 14 days after LLC cells injection. Mice were anesthetized and sacrificed 3 days after final administration. The volume of pleural effusion n was measured using 1 ml syringe. Micro blood vessel density (MVD), Lymphatic micro vessel density (LMVD), the expression level of vascular endothelial growth factor A (VEGF-A) and VEGF-C were observed by immunohistochemistry (IHC) staining. RESULTS: The volume of pleural effusion as well as the number of pleural tumor foci, MVD and the expression of VEGF-A were significantly reduced in high dose of Endostar treat group. More importantly, LMVD and the expression of VEGF-C were markedly lower in treat group than those in the other three control groups. CONCLUSION: Our work demonstrated that Endostar played an efficient anti-cancer role in MPE through its suppressive effect on angiogenesis and lymphangiogenesis, which provided a certain theoretical basis for the effectiveness of Endostar on the MPE treatment.


Assuntos
Carcinoma Pulmonar de Lewis/tratamento farmacológico , Endostatinas/farmacologia , Linfangiogênese/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Derrame Pleural Maligno/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Animais , Carcinoma Pulmonar de Lewis/complicações , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Endostatinas/uso terapêutico , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Linfangiogênese/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/etiologia , Derrame Pleural Maligno/complicações , Derrame Pleural Maligno/patologia , Proteínas Recombinantes/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA