Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomacromolecules ; 25(7): 4569-4580, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38869359

RESUMO

Acute myeloid leukemia (AML) is often associated with poor prognosis and survival. Small molecule inhibitors, though widening the treatment landscape, have limited monotherapy efficacy. The combination therapy, however, shows suboptimal clinical outcomes due to low bioavailability, overlapping systemic toxicity and drug resistance. Here, we report that CXCR4-mediated codelivery of the BCL-2 inhibitor venetoclax (VEN) and the FLT3 inhibitor sorafenib (SOR) via T22 peptide-tagged disulfide cross-linked polymeric micelles (TM) achieves synergistic treatment of FLT3-ITD AML. TM-VS with a VEN/SOR weight ratio of 1/4 and T22 peptide density of 20% exhibited an extraordinary inhibitory effect on CXCR4-overexpressing MV4-11 AML cells. TM-VS at a VEN/SOR dosage of 2.5/10 mg/kg remarkably reduced leukemia burden, prolonged mouse survival, and impeded bone loss in orthotopic MV4-11-bearing mice, outperforming the nontargeted M-VS and oral administration of free VEN/SOR. CXCR4-mediated codelivery of BCL-2 and FLT3 inhibitors has emerged as a prospective clinical treatment for FLT3-ITD AML.


Assuntos
Leucemia Mieloide Aguda , Proteínas Proto-Oncogênicas c-bcl-2 , Receptores CXCR4 , Sorafenibe , Sulfonamidas , Tirosina Quinase 3 Semelhante a fms , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/genética , Animais , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/administração & dosagem , Sorafenibe/farmacologia , Sorafenibe/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Micelas
2.
Biomacromolecules ; 25(7): 4440-4448, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38907698

RESUMO

Supramolecular delivery systems with the prolonged circulation, the potential for diverse functionalization, and few toxin-related limitations have been extensively studied. For the present study, we constructed a linear polyglycerol-shelled polymersome attached with the anti-HER-2-antibody trastuzumab. We then covalently loaded the anticancer drug DM1 in the polymersome via dynamic disulfide bonding. The resulted trastuzumab-polymersome-DM1 (Tra-PS-DM1) exhibits a mean size of 95.3 nm and remarkable drug loading efficiency % of 99.3%. In addition to its superior stability, we observed the rapid release of DM1 in a controlled manner under reductive conditions. Compared to the native polymersomes, Tra-PS-DM1 has shown greatly improved cellular uptake and significantly reduced IC50 up to 17-fold among HER-2-positive cancer cells. Moreover, Tra-PS-DM1 demonstrated superb growth inhibition of HER-2-positive tumoroids; specifically, BT474 tumoroids shrunk up to 62% after 12 h treatment. With exceptional stability and targetability, the PG-shelled Tra-PS-DM1 appears as an attractive approach for HER-2-positive tumor treatment.


Assuntos
Neoplasias da Mama , Glicerol , Polímeros , Receptor ErbB-2 , Trastuzumab , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Glicerol/química , Feminino , Polímeros/química , Trastuzumab/farmacologia , Trastuzumab/química , Trastuzumab/administração & dosagem , Receptor ErbB-2/metabolismo , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Ado-Trastuzumab Emtansina/farmacologia
3.
Biomater Sci ; 11(14): 4985-4994, 2023 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-37334506

RESUMO

Multiple myeloma (MM) is a neoplasm of aberrant plasma cells and ranks second among hematologic malignancies. Despite a substantial improvement in clinical outcomes with advances in therapeutic modalities over the past two decades, MM remains incurable, necessitating the development of new and potent therapies. Herein, we engineered a daratumumab-polymersome-DM1 conjugate (DPDC) based highly potent and CD38-selective immuno-nano-DM1 toxin for depleting MM cells in vivo. DPDC with controllable daratumumab density and disulfide-linked DM1 is of small size (51-56 nm), with high stability and reduction-triggered DM1 release. D6.2PDC potently inhibited the proliferation of CD38-overexpressed LP-1 and MM.1S MM cells with IC50 values of 2.7 and 1.2 ng DM1 equiv. per mL, about 4-fold stronger than non-targeted PDC. Moreover, D6.2PDC effectively and safely depleted LP-1-Luc MM cells in an orthotopic mouse model at a low DM1 dosage of 0.2 mg kg-1, thus alleviating osteolytic bone lesion and extending the median survival by 2.8-3.5-fold compared to all controls. This CD38-selective DPDC provides a safe and potent treatment strategy for MM.


Assuntos
Neoplasias Hematológicas , Mieloma Múltiplo , Camundongos , Animais , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , ADP-Ribosil Ciclase 1 , Linhagem Celular Tumoral
4.
Comput Intell Neurosci ; 2023: 8288527, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37284055

RESUMO

An online policy learning algorithm is used to solve the optimal control problem of the power battery state of charge (SOC) observer for the first time. The design of adaptive neural network (NN) optimal control is studied for the nonlinear power battery system based on a second-order (RC) equivalent circuit model. First, the unknown uncertainties of the system are approximated by NN, and a time-varying gain nonlinear state observer is designed to address the problem that the resistance capacitance voltage and SOC of the battery cannot be measured. Then, to realize the optimal control, a policy learning-based online algorithm is designed, where only the critic NN is required and the actor NN widely used in most design of the optimal control methods is removed. Finally, the effectiveness of the optimal control theory is verified by simulation.


Assuntos
Redes Neurais de Computação , Dinâmica não Linear , Algoritmos , Simulação por Computador , Fontes de Energia Elétrica
5.
Adv Mater ; 35(32): e2209984, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37321606

RESUMO

Acute myeloid leukemia (AML) is afflicted by a high-mortality rate and few treatment options. The lack of specific surface antigens severely hampers the development of targeted therapeutics and cell therapy. Here, it is shown that exogenous all-trans retinoic acid (ATRA) mediates selective and transient CD38 upregulation on leukemia cells by up to 20-fold, which enables high-efficiency targeted nanochemotherapy of leukemia with daratumumab antibody-directed polymersomal vincristine sulfate (DPV). Strikingly, treatment of two CD38-low expressing AML orthotopic models with ATRA and DPV portfolio strategies effectively eliminates circulating leukemia cells and leukemia invasion into bone marrow and organs, leading to exceptional survival benefits with 20-40% of mice becoming leukemia-free. The combination of exogenous CD38 upregulation and antibody-directed nanotherapeutics provides a unique and powerful targeted therapy for leukemia.


Assuntos
Leucemia Mieloide Aguda , Antineoplásicos/uso terapêutico , Regulação para Cima , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/imunologia , Anticorpos/uso terapêutico , Antígenos/imunologia , Humanos , Animais , Camundongos , ADP-Ribosil Ciclase 1/imunologia , Tretinoína/uso terapêutico
7.
J Control Release ; 350: 122-131, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35973474

RESUMO

Transarterial chemoembolization (TACE) with free doxorubicin-lipiodol emulsions (free DOX/L) is a favored clinical treatment for advanced hepatocellular carcinoma (HCC) patients ineligible for radical therapies; however, its inferior colloidal stability not only greatly reduces its tumor retention but also hastens drug release into blood circulation, leading to suboptimal clinical outcomes. Here, we find that disulfide-crosslinked polymersomes carrying doxorubicin (Ps-DOX) form super-stable and homogenous water-in-oil microemulsions with lipiodol (Ps-DOX/L). Ps-DOX/L microemulsions had tunable sizes ranging from 14 to 44 µm depending on the amount of Ps-DOX, were stable over 2 months storage as well as centrifugation, and exhibited nearly zero-order DOX release within 15 days. Of note, Ps-DOX induced 2.3-13.4 fold better inhibitory activity in all tested rat, murine and human liver tumor cells than free DOX likely due to its efficient redox-triggered intracellular drug release. Interestingly, transarterial administration of Ps-DOX/L microemulsions in orthotopic rat N1S1 syngeneic HCC model showed minimal systemic DOX exposure, high and long hepatic DOX retention, complete tumor elimination, effective inhibition of angiogenesis, and depleted adverse effects, significantly outperforming clinically used free DOX/L emulsions. This smart polymersome stabilization of doxorubicin-lipiodol microemulsions provides a novel TACE strategy for advanced tumors.


Assuntos
Carcinoma Hepatocelular , Quimioembolização Terapêutica , Neoplasias Hepáticas , Animais , Antibióticos Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Dissulfetos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Emulsões/uso terapêutico , Óleo Etiodado/uso terapêutico , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Ratos , Água
9.
Biomacromolecules ; 23(1): 377-387, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34913676

RESUMO

Acute lymphoblastic leukemia (ALL) is the most common malignancy in children. Although intensive chemotherapy greatly improved the survival rate, it is often accompanied by severe and lifelong side effects as a result of weak ALL selectivity. The intensive and poorly selective chemotherapy is also detrimental to patients' immune system. There is an urgent need to develop more selective and less toxic chemotherapy for ALL. Here, we report daratumumab-polymersome-vincristine (DP-VCR) as a CD38-directed nanotherapy for ALL. DP-VCR showed selective uptake in CD38-positive 697 and Nalm-6-Luc ALL cells and potent anti-ALL activity with an IC50 as low as 0.06 nM VCR, which was 13.7-fold more potent than free VCR. In contrast, no toxicity to human peripheral blood mononuclear cells was detected for DP-VCR even at 108.3 nM VCR. The apoptotic assays confirmed a high selectivity of DP-VCR to CD38-positive ALL cells. DP-VCR exhibited superior treatment of both 697 and Nalm-6-Luc orthotopic ALL models to all controls, as revealed by significant survival benefit and marked reduction of leukemia burden in bone marrow, blood, spleen, and liver. Importantly, DP-VCR induced few side effects. DP-VCR emerges as a safe and potent nanotherapy for CD38-positive ALL.


Assuntos
Leucócitos Mononucleares , Leucemia-Linfoma Linfoblástico de Células Precursoras , Contagem de Células , Criança , Humanos , Leucócitos Mononucleares/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Vincristina/farmacologia , Vincristina/uso terapêutico
10.
Biomacromolecules ; 23(1): 100-111, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34913340

RESUMO

Targeted nanomedicines particularly armed with monoclonal antibodies are considered to be the most promising advanced chemotherapy for malignant cancers; however, their development is hindered by their instability and drug leakage problems. Herein, we constructed a robust cetuximab-polymersome-mertansine nanodrug (C-P-DM1) for highly potent and targeted therapy of epidermal growth factor receptor (EGFR)-positive solid tumors. C-P-DM1 with a tailored cetuximab surface density of 2 per P-DM1 exhibited a size of ca. 60 nm, high stability with minimum DM1 leakage, glutathione-triggered release of native DM1, and 6.0-11.3-fold stronger cytotoxicity in EGFR-positive human breast (MDA-MB-231), lung (A549), and liver (SMMC-7721) cancer cells (IC50 = 27.1-135.5 nM) than P-DM1 control. Notably, intravenous injection of C-P-DM1 effectively repressed subcutaneous MDA-MB-231 breast cancer and orthotopic A549-Luc lung carcinoma in mice without inducing toxic effects. Strikingly, intratumoral injection of C-P-DM1 completely cured 60% of mice bearing breast tumor without recurrence. This robust cetuximab-polymersome-mertansine nanodrug provides a promising new strategy for targeted treatment of EGFR-positive solid malignancies.


Assuntos
Neoplasias da Mama , Cetuximab , Maitansina , Nanopartículas , Animais , Anticorpos Monoclonais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cetuximab/farmacologia , Receptores ErbB/metabolismo , Feminino , Humanos , Maitansina/farmacologia , Camundongos , Nanopartículas/metabolismo , Nanopartículas/uso terapêutico
11.
J Control Release ; 340: 331-341, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34774889

RESUMO

Antibody-drug conjugates (ADCs) are among the most significant advances in clinical cancer treatments, however, they are haunted with fundamental issues like low drug/antibody ratio (DAR), need of large amount of antibody, and complex chemistry. Targeted nanomedicines while offering a promising alternative to ADCs are afflicted with drug leakage and inferior cancer-specificity. Herein, we developed an intelligent cell-selective nanotoxin based on anti-CD44 antibody-polymersome-DM1 conjugates (aCD44-AP-DM1) for potent treatment of solid tumors. DM1 was simultaneously coupled to vesicular membrane via disulfide bonds during self-assembly and anti-CD44 antibody was facilely clicked onto polymersome surface, tailor-making an optimal aCD44-AP-DM1 with a controlled antibody density of 5.0, extraordinary DAR of 275, zero drug leakage and rapid reduction-responsive DM1 release. aCD44-AP-DM1 displayed a high specificity and exceptional cytotoxicity toward MDA-MB-231 triple negative breast cancer, SMMC-7721 hepatocellular carcinoma and A549 non-small cell lung cancer cells with half-maximal inhibitory concentrations (IC50) of 21.4, 3.7 and 64.6 ng/mL, respectively, 3.6-47.2-fold exceeding non-targeted P-DM1. Intriguingly, the systemic administration of aCD44-AP-DM1 significantly suppressed subcutaneous MDA-MB-231 tumor xenografts in nude mice while intratumoral injection achieved complete tumor eradication in four out of five mice, without causing toxicity. This intelligent cell-selective nanotoxin has emerged as a better platform over ADCs for targeted cancer therapy.


Assuntos
Neoplasias da Mama , Carcinoma Pulmonar de Células não Pequenas , Imunoconjugados , Neoplasias Pulmonares , Maitansina , Neoplasias de Mama Triplo Negativas , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Trastuzumab , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
12.
Adv Mater ; 33(39): e2007787, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34369013

RESUMO

Multiple myeloma (MM) is a second ranking hematological malignancy. Despite the fast advancement of new treatments such as bortezormib and daratumumab, MM patients remain incurable and tend to eventually become relapsed and drug-resistant. Development of novel therapies capable of depleting MM cells is strongly needed. Here, daratumumab immunopolymersomes carrying vincristine sulfate (Dar-IPs-VCR) are reported for safe and high-efficacy CD38-targeted chemotherapy and depletion of orthotopic MM in vivo. Dar-IPs-VCR made by postmodification via strain-promoted click reaction holds tailored antibody density (2.2, 4.4 to 8.7 Dar per IPs), superb stability, small size (43-49 nm), efficacious VCR loading, and glutathione-responsive VCR release. Dar4.4 -IPs-VCR induces exceptional anti-MM activity with an IC50 of 76 × 10-12 m to CD38-positive LP-1 MM cells, 12- and 20-fold enhancement over nontargeted Ps-VCR and free VCR controls, respectively. Intriguingly, mice bearing orthotopic LP-1-Luc MM following four cycles of i.v. administration of Dar4.4 -IPs-VCR at 0.25 mg VCR equiv. kg-1 reveal complete depletion of LP-1-Luc cells, superior survival rate to all controls, and no body weight loss. The bone and histological analyses indicate bare bone and organ damage. Dar-IPs-VCR appears as a safe and targeted treatment for CD38-overexpressed hematological malignancies.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Monoclonais/imunologia , Antineoplásicos Fitogênicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , ADP-Ribosil Ciclase 1/metabolismo , Animais , Anticorpos Monoclonais/química , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Nanomedicina , Nanoestruturas/química , Tamanho da Partícula , Tíbia/patologia , Transplante Heterólogo , Vincristina/química
14.
J Mater Chem B ; 8(31): 6866-6876, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32249886

RESUMO

Engineering nanoparticles (NPs) with multifunctionality has become a promising strategy for cancer theranostics. Herein, theranostic polymer NPs are fabricated via the assembly of amphiphilic paramagnetic block copolymers (PCL-b-PIEtMn), in which IR-780 and doxorubicin (DOX) were co-encapsulated, for magnetic resonance (MR) and near infrared fluorescence (NIRF) imaging as well as for photo thermal therapy (PTT)-enhanced chemotherapy. The synthesized amphiphilic paramagnetic block copolymers demonstrated high relaxivity (r1 = 7.05 mM-1 s-1). The encapsulated DOX could be released with the trigger of near infrared (NIR) light. In vivo imaging confirmed that the paramagnetic NPs could be accumulated effectively at the tumor sites. Upon the NIR laser irradiation, tumor growth was inhibited by PTT-enhanced chemotherapy. The advantages of the reported system lie in the one-step convergence of multiple functions (i.e., imaging and therapy agents) into a one delivery vehicle and the dual mode imaging-guided synergistic PTT and chemotherapy. This study represents a new drug delivery vehicle of paramagnetic NPs for visualized theranostics.


Assuntos
Interações Hidrofóbicas e Hidrofílicas , Imãs/química , Polímeros/química , Polímeros/uso terapêutico , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Humanos , Células MCF-7 , Nanopartículas/química , Imagem Óptica , Fototerapia
15.
ACS Macro Lett ; 9(9): 1292-1302, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35638634

RESUMO

The development of smart polymer vehicles to carry and release cytotoxic drugs to tumor tissues and cells while reducing the exposure of drugs in the blood and healthy organs is a highly challenging task with continuously growing interest from multiple fields, including polymer science, pharmaceutical science, nanotechnology, and clinical oncology. Inspired by the unique tumor microenvironment, such as mild acidity and overexpressed enzymes, functional polymer prodrugs and nanoparticles with reversible charge, detachable PEG shell, activatable ligand, and switchable size have been designed to enhance tumor deposition, tumor penetration, tumor cell uptake, and tumoral drug release. Utilizing biological signals inside tumor cells, such as acidic endo/lysosomal pH, elevated glutathione levels, and reactive oxygen species, responsive polymer prodrugs and nanoparticles with good extracellular stability but fast intracellular disintegration have been engineered for specific intracellular drug release. These biological stimuli-sensitive polymer prodrugs and nanoparticles have shown superior specificity and therapeutic efficacy to nonsensitive counterparts and, in certain cases, even clinically approved systems in varying tumor models. In this Viewpoint, design strategies and recent advances of biological stimuli-responsive polymer prodrugs and nanoparticles for tumor-specific drug delivery will be highlighted, and their challenges and future perspectives will be discussed.

16.
Biomacromolecules ; 21(1): 104-113, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31532629

RESUMO

Metastasis is responsible for >90% of the deaths of breast cancer patients in the clinic. Here, we report on cross-linked multifunctional hyaluronic acid nanoparticles carrying docetaxel (DTX-CMHN) for enhanced suppression of highly metastatic 4T1 breast tumors in vivo. DTX-CMHN was formed from a single and all-natural hyaluronic acid-g-polytyrosine-lipoic acid conjugate (HA-g-PTyr-LA; HA, 20 kDa; PTyr, 2.2 kDa), and the size of DTX-CMHN increased from 69 to 78 to 96 nm as the increasing degree of substitution (DS) of PTyr increased from 4 to 11 to 15, respectively. Robust encapsulation of DTX was obtained when DS ≥ 11. DTX-CMHN while steady in a nonreducing environment was destabilized under 10 mM glutathione releasing ∼90% of the DTX within 24 h. It is noteworthy that DTX-CMHN exhibited better antitumor, antimigration, and anti-invasion activity in CD44-overexpressed 4T1-Luc breast cancer cells than free DTX. Interestingly, DTX-CMHN displayed a long elimination half-life of 5.75 h, in contrast to half-lives of 2.11 and 0.75 h for its non-cross-linked counterpart (DTX-MHN) and free DTX, respectively. In vivo therapeutic studies showed significantly better inhibition of primary 4T1-Luc tumor growth and lung metastasis and lower toxicity of DTX-CMHN compared with that of free DTX. These multifunctional nanoformulations based on a single and all-natural hyaluronic acid conjugate emerge as a potential nanoplatform for targeted treatment of CD44-positive metastatic tumors.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Receptores de Hialuronatos , Nanopartículas/química , Animais , Antineoplásicos/farmacocinética , Neoplasias da Mama/patologia , Docetaxel/administração & dosagem , Docetaxel/farmacocinética , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Ácido Hialurônico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Terapia de Alvo Molecular/métodos , Nanomedicina/métodos , Nanopartículas/administração & dosagem , Ácido Tióctico/química , Distribuição Tecidual , Tirosina/química , Ensaios Antitumorais Modelo de Xenoenxerto
18.
ACS Appl Mater Interfaces ; 11(50): 46548-46557, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31763810

RESUMO

Actively targeted nanomedicines have promised to revolutionize cancer treatment; however, their clinical translation has been limited by either low targetability, use of unsafe materials, or tedious fabrication. Here, we developed CD44 and folate receptor (FR) dually targeted nanoparticulate doxorubicin (HA/FA-NP-DOX) based on a direct conjugate of two purely natural ligands, hyaluronic acid and folic acid (FA), for safe, highly specific, and potent treatment of ovarian tumors in vivo. HA/FA-NP-DOX had a small size and high DOX loading, wherein the particle size decreased from 115, 93, to 89 nm with increasing degree of substitution of FA from 6.4, 8.5, to 11.1, while increased from 80, 93, to 103 nm with increasing DOX loading from 15.0, 23.1, to 31.4 wt %. Interestingly, HA/FA-NP-DOX exhibited excellent lyophilization redispersibility and long-term storage stability with negligible drug leakage while it released 91% of DOX in 48 h at pH 5.0. Cellular studies corroborated that HA/FA-NP-DOX possessed high selectivity to both CD44 and FR, resulting in strong killing of CD44- and FR-positive SKOV-3 ovarian cancer cells while low toxicity against CD44- and FR-negative L929 fibroblast cells. In vivo studies revealed a long elimination half-life of 5.6 h, an elevated tumor accumulation of 12.0% ID/g, and an effective inhibition of the SKOV-3 ovarian tumor for HA/FA-NP-DOX, leading to significant survival benefits over free DOX·HCl and phosphate-buffered saline controls. These dually targeted nanomedicines are simple and safe, providing a potentially translatable treatment for CD44- and FR-positive malignancies.


Assuntos
Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias Ovarianas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Doxorrubicina/química , Feminino , Receptor 1 de Folato/antagonistas & inibidores , Receptor 1 de Folato/genética , Humanos , Receptores de Hialuronatos/antagonistas & inibidores , Receptores de Hialuronatos/genética , Ligantes , Camundongos , Nanomedicina/métodos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Biomacromolecules ; 20(12): 4299-4311, 2019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31659901

RESUMO

Nanomedicines are deemed as the most promising treatment modality for malignant cancers. Particularly, cancer nanomedicines based on synthetic polypeptides have gained interest because they possess excellent safety, unique hierarchical structure, and tailorable functionalities to suit for delivery of diverse drugs including synthetic drugs, peptides, proteins, and nucleic acids. A few polypeptide-based nanoformulations (e.g., NK105, NC6004, NK911, CT2103) are under phases I-III clinical investigation for treating patients with advanced solid tumors. In recent years, progress has been made in the development of robust and high drug loading, tumor-targeting, membrane-disrupting, and stimuli-sensitive nanomedicines from de novo functional polypeptides, which afford not only better safety and reduced adverse effects, but also further improved anticancer efficacy over clinical formulations. Moreover, virus-mimicking vehicles have been devised from polypeptides for efficient nonviral delivery of highly potent peptides, proteins, and nucleic acids, greatly advancing biotherapy for cancers. In this Perspective, we highlight the state-of-the-art design and fabrication of cancer nanomedicines based on synthetic polypeptides and, at the end, give our viewpoints on their future development for targeted cancer therapy and potential challenges for clinical translation.


Assuntos
Antineoplásicos , Sistemas de Liberação de Medicamentos , Nanomedicina , Nanopartículas , Neoplasias , Peptídeos , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Peptídeos/síntese química , Peptídeos/química , Peptídeos/uso terapêutico
20.
Biomacromolecules ; 20(10): 3855-3863, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31513391

RESUMO

Monoclonal antibodies can effectively target to tumors in patients, as validated by antibody-drug conjugates (ADCs). The clinically used ADCs, nevertheless, are restricted to toxins only and suffer from low drug content, excessive use of antibody, and high cost. Here, we report on trastuzumab-decorated disulfide-cross-linked polymersomes (Tra-Ps) for specific delivery of epirubicin hydrochloride (EPI·HCl) to HER2-positive SKOV-3 ovarian tumor. EPI·HCl-loaded Tra-Ps (Tra-Ps-EPI) with a small size of 50-60 nm and varying Tra surface densities (0.5 to 2.4 Tra per Ps) were conveniently obtained via post-conjugation of thiolated trastuzumab onto the surface of maleimide-functionalized Ps-EPI with a drug loading content of 12.7 wt %. Interestingly, Tra-Ps with 1.3 trastuzumab on the surface exhibited a 6-fold higher binding affinity to the HER2 extracellular domain than that of native trastuzumab. In vitro studies revealed that Tra-Ps-EPI with long-term storage stability could rapidly release drugs under a reductive condition and efficiently deliver a large amount of EPI·HCl to HER2-positive SKOV-3 cells, leading to stronger cytotoxicity than the nontargeted Ps-EPI. Moreover, Tra-Ps-EPI displayed a long circulation time (ca. 8 h), deep tumor penetration, and superior tumor growth inhibition in SKOV-3 ovarian tumor-bearing nude mice, which were more effective than free EPI·HCl and nontargeted Ps-EPI. These HER2-specific reduction-sensitive immunopolymersomes with high loading of epirubicin emerge as an attractive treatment for HER2-positive tumors.


Assuntos
Epirubicina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Receptor ErbB-2/genética , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Imunoconjugados/farmacologia , Camundongos , Camundongos Nus , Neoplasias Ovarianas/genética , Trastuzumab/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...