Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Nano Lett ; 23(18): 8593-8601, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37625135

RESUMO

Despite the great progress of current bacterially based biotherapeutics, their unsatisfying efficacy and underlying safety problems have limited their clinical application. Herein, inspired by probiotic Escherichia coli strain Nissle 1917, probiotic-derived outer membrane vesicles (OMVs) are found to serve as an effective therapeutic platform for the treatment of inflammatory bowel disease (IBD). To further enhance the therapeutic effect, the probiotic-derived OMV-encapsulating manganese dioxide nanozymes are constructed, named nanoprobiotics, which can adhere to inflamed colonic epithelium and eliminate intestinal excess reactive oxygen species in the murine IBD model. Moreover, combined with the anti-inflammatory medicine metformin, nanoprobiotics could further remold the pro-inflammatory microenvironment, improve the overall richness and diversity of the gut microbiota, and exhibit better therapeutic efficacy than commercial IBD chemotherapeutics. Importantly, insignificant overt systemic toxicity in this treatment was observed. By integrating cytokine storm calm with biotherapy, we develop a safe and effective bionanoplatform for the effective treatment of inflammation-mediated intestinal diseases.

2.
Mol Pharm ; 20(5): 2642-2649, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37043673

RESUMO

Liposomes (lipos), one of the most successful nanotherapeutics in the clinic, have made a rapid advance over the past few years. However, still, several challenges exist for lipos for clinical practice, such as low drug loading and premature drug leakage during in vivo circulation. Paclitaxel (PTX), a commonly used first-line drug for cancer chemotherapy, was chosen as the model drug. Due to its non-ionizable and water-insoluble characteristics, the drug-loading efficiency of the marketable PTX lipos, Lipusu, is only 6.76%. Herein, we designed an ionizable PTX prodrug (PTXP) by modifying phenylboronic acid on the C2' hydroxyl group of PTX for the remote loading of liposomal formulations through the pH gradient method. Compared with Lipusu, PTXP lipos displayed a 34% higher loading efficiency and an encapsulation efficiency of approximately 95%. A series of in vitro/vivo experiments indicated that PTXP lipos possess colloidal stability, prolonged blood circulation, high tumor site accumulation, potent anti-tumor effects, and safety. A combination of ionizable prodrugs and remote loading has proved to be an effective and simple strategy to achieve high liposomal encapsulation efficiency of poorly soluble non-ionizable drugs for clinical application.


Assuntos
Neoplasias da Mama , Pró-Fármacos , Humanos , Feminino , Lipossomos , Linhagem Celular Tumoral , Paclitaxel/uso terapêutico , Neoplasias da Mama/tratamento farmacológico
3.
Nano Lett ; 22(12): 5055-5064, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35583490

RESUMO

Oncolytic viruses (OVs) have been widely used as anticancer therapeutics because of their systemic immune responses during viral replication. However, the low enrichment of OVs within tumors and limited immune activation have hindered their clinical application. Herein, we proposed the concept of bacteria-assisted targeting of OVs to tumors, with liposome-cloaked oncolytic adenoviruses (OAs) conjugated onto tumor-homing Escherichia coli BL21 (designated as E. coli-lipo-OAs) for enhanced cancer immunotherapy. Notably, the enrichment of OAs transported by self-propelled bacterial microbe vehicles in E. coli-lipo-OAs in a nonsmall cell lung tumor can be potentiated by more than 170-fold compared with that of intravenously injected bare OAs. In vivo studies further revealed that E. coli-lipo-OAs administered intravenously significantly enhanced antitumor immunity through bacterial-viral-augmented immune responses. Our findings suggest that the self-driving microbe vehicle as a systemic delivery system for OVs can be a potent platform for developing future anticancer biotherapeutics at the clinical level.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Escherichia coli , Humanos , Imunoterapia , Neoplasias/terapia , Vírus Oncolíticos/genética
4.
Small ; 16(45): e2005039, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33078579

RESUMO

The current state of chemotherapy is far from satisfaction, restricted by the inefficient drug delivery and the off-target toxicity. Prodrug nanoassemblies are emerging as efficient platforms for chemotherapy. Herein, three docetaxel dimeric prodrugs are designed using diselenide bond, disulfide bond, or dicarbide bond as linkages. Interestingly, diselenide bond-bridged dimeric prodrug can self-assemble into stable nanoparticles with impressive high drug loading (≈70%, w/w). Compared with disulfide bond and dicarbide bond, diselenide bond greatly facilitates the self-assembly of dimeric prodrug, and then improves the colloidal stability, blood circulation time, and antitumor efficacy of prodrug nanoassemblies. Furthermore, the redox-sensitive diselenide bond can specifically respond to the overexpressed reactive oxygen species and glutathione in tumor cells, leading to tumor-specific drug release. Therefore, diselenide bond bridged prodrug nanoassemblies exhibit discriminating cytotoxicity between tumor cells and normal cells, significantly alleviating the systemic toxicity of docetaxel. The present work gains in-depth insight into the impact of diselenide bond on the dimeric prodrug nanoassemblies, and provides promising strategies for the rational design of the high efficiency-low toxicity chemotherapeutical nanomedicines.


Assuntos
Antineoplásicos , Nanopartículas , Pró-Fármacos , Linhagem Celular Tumoral , Docetaxel , Liberação Controlada de Fármacos , Nanomedicina
5.
Mol Pharm ; 15(8): 3121-3132, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29979604

RESUMO

Transdermal delivery is one of important controlled drug release strategies for drug development. Cubic phases are the assemblies of amphiphilic molecules in water with the hydrophilic-hydrophobic interpenetrating network for transdermal delivery of both hydrophilic and hydrophobic drugs. However, many details about the transdermal delivery of drugs from cubic phases remain unclear. Here, metformin hydrochloride (Met) cubic phases were prepared with glyceryl monooleate (GMO), ethanol, and water. The cubic structure was identified with the polarizing light microscopy and small-angle X-ray scattering method. Dissipative particle dynamics (DPD) was used for building the microstructures of the cubic phases to explore the mechanism of drug release that mainly depended on drug diffusion from the water channels of cubic phases in accordance with the Higuchi equation of in vitro release experiments. The coarse-grained model and molecular docking method showed that GMO could enhance drug permeation through the skin by disturbing the interaction between Met and the skin proteins, and increasing the fluidity of skin lipids, which was confirmed with the Fourier transform infrared spectroscopy, Langmuir monolayer, and immunohistochemistry. Furthermore, in vitro permeation experiments showed the high Met transdermal improvement of cubic phases. Cubic phases are an ideal transdermal delivery system of Met. In silico methods are very useful for analyzing the molecular mechanisms of transdermal formulations.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Metformina/administração & dosagem , Adesivo Transdérmico , Administração Cutânea , Animais , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Etanol/química , Glicerídeos/química , Interações Hidrofóbicas e Hidrofílicas , Masculino , Metformina/química , Metformina/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Pele/efeitos dos fármacos , Água/química , Perda Insensível de Água/efeitos dos fármacos
6.
Bioconjug Chem ; 27(5): 1360-72, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27070505

RESUMO

Citronellol-cabazitaxel (CIT-ss-CTX) conjugate self-assembled nanoparticles (CSNPs) were designed and prepared by conjugating cabazitaxel with citronellol via the disulfide bond that is redox-sensitive to the high concentration of glutathione within tumor cells. Notably, the CSNPs maintained in the cell cytotoxicity. Moreover, the AUC0-t of CSNPs was 6.5-fold higher than that of cabazitaxel solutions and the t1/2 was prolonged 2.3 times. Furthermore, we found that CSNPs could be employed as an efficient carrier for other hydrophobic drugs or imaging agents. Thus, the in vivo targeting study was implemented via using 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyanine iodide (DiR)-loaded CSNPs as imaging agent, which showed CSNPs could effectively accumulate at the tumor site. Curcumin, a hydrophobic anticancer drug, was successfully loaded in CSNPs which exhibits good stability and synergistic antitumor effects. The citronellol-cabazitaxel conjugate therefore has a promising perspective as a multifunctional nanomedicine for combination therapy and theranostics attributed to its long-circulation property, redox-sensitive mechanism, and high drug coloading capability.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Portadores de Fármacos/química , Monoterpenos/química , Nanomedicina , Taxoides/química , Taxoides/farmacologia , Monoterpenos Acíclicos , Animais , Transporte Biológico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Estabilidade de Medicamentos , Humanos , Monoterpenos/metabolismo , Monoterpenos/farmacocinética , Nanopartículas/química , Oxirredução , Ratos , Distribuição Tecidual
7.
Biomed Pharmacother ; 177: 117063, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38968800

RESUMO

Off-targeting toxicity and immunosuppressive tumor microenvironment still restrict the therapeutic requirement of photodynamic therapy (PDT). The development of metal ion-coordination-based nanoparticles (NPs) for cancer therapy has advantages, such as precious nanostructure and potent therapeutic effect as well as great safety. In this study, we prepared calcium ions (Ca2+)-coordination photosensitizer NPs, based on Ca2+-pyrochloric acid (PPA)-coordination as the new photosensitive nanoamplifiers, and microneedles (MNs) as the personalized apparatus, and investigated the nanoamplifiers for treating the melanoma via transdermal administration. This nanoamplifiers was synthesized via a simple coordination of Ca2+ and PPA with the addition of bovine serum albumin (BSA), and further fabricated into MNs (nanoamplifiers@MNs). Following inserted into the tumor, the released nanoamplifiers from the tips and back layer exhibited great photodynamic activity under irradiation, inducing cancer cell death. Meanwhile, Ca2+ acted as the second messenger, promoting M1 polarization of macrophages and maturation of dendritic cells (DCs), thereby enhancing the immune activation effect in the tumor microenvironment. As a result, such nanoamplifiers effectively achieved significant efficacy against malignant melanoma tumors by synergistically tumor killing and potent anti-tumor immune activation without obviously side effect. This work demonstrated the potential of MNs-mediated metal ion-coordination-based nanoamplifier as a novel photodynamic therapeutic platform for the efficient and safe treatment of cancer.

8.
J Control Release ; 365: 668-687, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042376

RESUMO

Anti-cancer therapeutics have achieved significant advances due to the emergence of immunotherapies that rely on the identification of tumors by the patients' immune system and subsequent tumor eradication. However, tumor cells often escape immunity, leading to poor responsiveness and easy tolerance to immunotherapy. Thus, the potentiated anti-tumor immunity in patients resistant to immunotherapies remains a challenge. Reactive oxygen species-based dynamic nanotherapeutics are not new in the anti-tumor field, but their potential as immunomodulators has only been demonstrated in recent years. Dynamic nanotherapeutics can distinctly enhance anti-tumor immune response, which derives the concept of the dynamic immuno-nanomedicines (DINMs). This review describes the pivotal role of DINMs in cancer immunotherapy and provides an overview of the clinical realities of DINMs. The preclinical development of emerging DINMs is also outlined. Moreover, strategies to synergize the antitumor immunity by DINMs in combination with other immunologic agents are summarized. Last but not least, the challenges and opportunities related to DINMs-mediated immune responses are also discussed.


Assuntos
Neoplasias , Humanos , Neoplasias/terapia , Oncologia , Fatores Imunológicos/uso terapêutico , Imunoterapia , Adjuvantes Imunológicos/uso terapêutico
9.
Adv Mater ; 36(7): e2306488, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37844257

RESUMO

Because the tumor-draining lymph nodes (TDLNs) microenvironment is commonly immunosuppressive, oncolytic microbe-induced tumor antigens aren't sufficiently cross-primed tumor specific T cells through antigen-presenting cells (e.g., dendritic cells (DCs)) in TDLNs. Herein, this work develops the micro-to-nano oncolytic microbial therapeutics based on pyranose oxidase (P2 O) overexpressed Escherichia coli (EcP) which are simultaneously encapsulated by PEGylated mannose and low-concentrated photosensitizer nanoparticles (NPs). Following administration, P2 O from this system generates toxic hydrogen peroxide for tumor regression and leads to the release of tumor antigens. The "microscale" EcP is triggered, following exposure to the laser irradiation, to secrete the "nanoscale" bacterial outer membrane vesicles (OMVs). The enhanced TDLNs delivery via OMVs significantly regulates the TDLNs immunomicroenvironment, promoting the maturation of DCs to potentiate tumor antigen-specific T cells immune response. The micro-to-nano oncolytic microbe is leveraged to exert tumor killing and remold TDLNs for initiating potent activation of DCs, providing promising strategies to facilitate microbial cancer vaccination.


Assuntos
Neoplasias , Humanos , Imunoterapia , Antígenos de Neoplasias , Células Dendríticas , Linfonodos , Microambiente Tumoral
10.
J Control Release ; 370: 653-676, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38735396

RESUMO

Pyroptosis, a non-apoptotic programmed cellular inflammatory death mechanism characterized by gasdermin (GSDM) family proteins, has gathered significant attention in the cancer treatment. However, the alarming clinical trial data indicates that pyroptosis-mediated cancer therapeutic efficiency is still unsatisfactory. It is essential to integrate the burgeoning biomedical findings and innovations with potent technology to hasten the development of pyroptosis-based antitumor drugs. Considering the rapid development of pyroptosis-driven cancer nanotherapeutics, here we aim to summarize the recent advances in this field at the intersection of pyroptosis and nanotechnology. First, the foundation of pyroptosis-based nanomedicines (NMs) is outlined to illustrate the reliability and effectiveness for the treatment of tumor. Next, the emerging nanotherapeutics designed to induce pyroptosis are overviewed. Moreover, the cross-talk between pyroptosis and other cell death modalities are discussed, aiming to explore the mechanistic level relationships to provide guidance strategies for the combination of different types of antitumor drugs. Last but not least, the opportunities and challenges of employing pyroptosis-based NMs in potential clinical cancer therapy are highlighted.


Assuntos
Antineoplásicos , Neoplasias , Piroptose , Piroptose/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Neoplasias/metabolismo , Neoplasias/patologia , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Animais , Nanomedicina/métodos , Nanotecnologia/métodos , Nanopartículas/administração & dosagem
11.
Nat Commun ; 14(1): 2933, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37217527

RESUMO

Oncolytic adenovirus (Ad) infection promotes intracellular autophagy in tumors. This could kill cancer cells and contribute to Ads-mediated anticancer immunity. However, the low intratumoral content of intravenously delivered Ads could be insufficient to efficiently activate tumor over-autophagy. Herein, we report bacterial outer membrane vesicles (OMVs)-encapsulating Ads as microbial nanocomposites that are engineered for autophagy-cascade-augmented immunotherapy. Biomineral shells cover the surface antigens of OMVs to slow their clearance during in vivo circulation, enhancing intratumoral accumulation. After entering tumor cells, there is excessive H2O2 accumulation through the catalytic effect of overexpressed pyranose oxidase (P2O) from microbial nanocomposite. This increases oxidative stress levels and triggers tumor autophagy. The autophagy-induced autophagosomes further promote Ads replication in infected tumor cells, leading to Ads-overactivated autophagy. Moreover, OMVs are powerful immunostimulants for remolding the immunosuppressive tumor microenvironment, facilitating antitumor immune response in preclinical cancer models in female mice. Therefore, the present autophagy-cascade-boosted immunotherapeutic method can expand OVs-based immunotherapy.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Feminino , Animais , Camundongos , Adenoviridae/genética , Membrana Externa Bacteriana , Peróxido de Hidrogênio , Neoplasias/patologia , Autofagia/fisiologia , Vírus Oncolíticos/genética , Microambiente Tumoral
12.
ACS Nano ; 17(15): 14461-14474, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37367941

RESUMO

Intravenous administration of oncolytic adenoviruses (OVs) is a hopeful tumor therapeutic modality. However, the sharp clearance of OVs by the immune system dampens its effectiveness. Many studies have attempted to extend the circulation of intravenously administered OVs, almost all by preventing OVs from binding to neutralizing antibodies and complements in the blood, but the results have not been satisfactory. In contrast to previous conclusions, we found that the key to improving the circulation of OVs is to prevent the formation of the virus-protein corona rather than simply preventing the binding of neutralizing antibodies or complements to OVs. After identifying the key protein components of the virus-protein corona, we proposed a virus-protein corona replacement strategy, where an artificial virus-protein corona was formed on OVs to completely prevent the interaction of OVs with key virus-protein corona components in the plasma. It was found that this strategy dramatically prolonged the circulation time of OVs by over 30 fold and increased the distribution of OVs in tumors by over 10-fold, resulting in superior antitumor efficacy in primary and metastatic tumor models. Our finding provides a perspective on intravenous delivery of OVs, shifting the focus of future studies from preventing OV binding with neutralization antibodies and complements to preventing OVs from interacting with key virus-protein corona components in the plasma.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Coroa de Proteína , Humanos , Vírus Oncolíticos/genética , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Neoplasias/terapia , Anticorpos Neutralizantes
13.
Biomed Pharmacother ; 167: 115577, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37757494

RESUMO

Gut microbiota can coordinate with different tissues and organs to maintain human health, which derives the concept of the gut-X axis. Conversely, the dysbiosis of gut microbiota leads to the occurrence and development of various diseases, such as neurological diseases, liver diseases, and even cancers. Therefore, the modulation of gut microbiota offers new opportunities in the field of medicines. Antibiotics, probiotics or other treatments might restore unbalanced gut microbiota, which effects do not match what people have expected. Recently, nanomedicines with the high targeting ability and reduced toxicity make them an appreciative choice for relieving disease through targeting gut-X axis. Considering this paradigm-setting trend, the current review summarizes the advancements in gut microbiota and its related nanomedicines. Specifically, this article introduces the immunological effects of gut microbiota, summarizes the gut-X axis-associated diseases, and highlights the nanotherapeutics-mediated treatment via remolding the gut-X axis. Moreover, this review also discusses the challenges in studies related to nanomedicines targeting the gut microbiota and offers the future perspective, thereby aiming at charting a course toward clinic.

14.
Mater Today Bio ; 20: 100644, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37214549

RESUMO

Self-assembled short peptides have intrigued scientists due to the convenience of synthesis, good biocompatibility, low toxicity, inherent biodegradability and fast response to change in the physiological environment. Therefore, it is necessary to present a comprehensive summary of the recent advances in the last decade regarding the construction, route of administration and application of self-assembled short peptides based on the knowledge on their unique and specific ability of self-assembly. Herein, we firstly explored the molecular mechanisms of self-assembly of short peptides, such as non-modified amino acids, as well as Fmoc-modified, N-functionalized, and C-functionalized peptides. Next, cell penetration, fusion, and peptide targeting in peptide-based drug delivery were characterized. Then, the common administration routes and the potential pharmaceutical applications (drug delivery, antibacterial activity, stabilizers, imaging agents, and applications in bioengineering) of peptide drugs were respectively summarized. Last but not least, some general conclusions and future perspectives in the relevant fields were briefly listed. Although with certain challenges, great opportunities are offered by self-assembled short peptides to the fascinating area of drug development.

15.
Adv Mater ; 35(28): e2212210, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37002917

RESUMO

Despite the superior tumor lytic efficacy of oncolytic viruses (OVs), their systemic delivery still faces the challenges of limited circulating periods, poor tumor tropism, and spontaneous antiviral immune responses. Herein, a virus-concealed tumor-targeting strategy enabling OVs' delivery toward lung metastasis via systemic administration is described. The OVs can actively infect, be internalized, and cloak into tumor cells. Then the tumor cells are subsequently treated with liquid-nitrogen-shocking to eliminate the pathogenicity. Such a Trojan Horse-like vehicle avoids virus neutralization and clearance in the bloodstream and facilitates tumor-targeted delivery for more than 110-fold virus enrichment in the tumor metastasis. In addition, this strategy can serve as a tumor vaccine and initiate endogenous adaptive antitumor effects through increasing the memory T cells and modulating the tumor immune microenvironment, including reducing the M2 macrophage, downregulating Treg cells, and priming T cells.


Assuntos
Vacinas Anticâncer , Neoplasias Pulmonares , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Vírus Oncolíticos/fisiologia , Neoplasias/terapia , Neoplasias Pulmonares/terapia , Microambiente Tumoral , Imunoterapia
16.
Nano Res ; 15(5): 4137-4153, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35194488

RESUMO

Oncolytic virotherapy (OVT) is a novel type of immunotherapy that induces anti-tumor responses through selective self-replication within cancer cells and oncolytic virus (OV)-mediated immunostimulation. Notably, talimogene laherparepvec (T-Vec) developed by the Amgen company in 2015, is the first FDA-approved OV product to be administered via intratumoral injection and has been the most successful OVT treatment. However, the systemic administration of OVs still faces huge challenges, including in vivo pre-existing neutralizing antibodies and poor targeting delivery efficacy. Recently, state-of-the-art progress has been made in the development of systemic delivery of OVs, which demonstrates a promising step toward broadening the scope of cancer immunotherapy and improving the clinical efficacy of OV delivery. Herein, this review describes the general characteristics of OVs, focusing on the action mechanisms of OVs as well as the advantages and disadvantages of OVT. The emerging multiple systemic administration approaches of OVs are summarized in the past five years. In addition, the combination treatments between OVT and traditional therapies (chemotherapy, thermotherapy, immunotherapy, and radiotherapy, etc.) are highlighted. Last but not least, the future prospects and challenges of OVT are also discussed, with the aim of facilitating medical researchers to extensively apply the OVT in the cancer therapy.

17.
Cancer Lett ; 551: 215934, 2022 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-36191678

RESUMO

Cancer immunotherapy (CIT) has revolutionized cancer treatment. However, the application of CIT is limited by low response rates and significant individual differences owing to a deficit in 1) immune recognition and 2) immune effector function. Extracellular vesicles (EVs) are cell-derived lipid bilayer-enclosed vesicles that mediate intercellular communication. The specific structure and content of EVs allows for multi-functional modulation of tumor immunity. Given their high biocompatibility, homologous targeting, and permeability across biological barriers, EVs have been evaluated as ideal carriers for promoting the efficacy and specificity of CIT. Herein, we first discuss the role of EVs in regulating tumor immunity and focus on the advantages of using EVs as a therapeutic tool for cancer treatment from a clinical perspective. Further, we outline the current progress in the development of biohybrid EVs for CIT and multi-functional EV-based strategies for overcoming the deficits in tumor immunity. Finally, we discuss the challenges associated with EV-based CIT and future perspectives in the context of ongoing clinical trials involving EV-based therapies, thus offering valuable insights into the future of multi-functional EVs in CIT.

18.
Adv Sci (Weinh) ; : e2205247, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36453573

RESUMO

The strategy of combining immune checkpoint inhibitors (ICIs) with anthracycline is recommended by clinical guidelines for the standard-of-care treatment of triple-negative breast cancer (TNBC). Nevertheless, several fundamental clinical principles are yet to be elucidated to achieve a great therapeutic effect, including cancer-homing delivery efficiency and ordinal-interval regime. Tumor-derived extracellular vesicles (TDEVs), as vectors for intratumoral intercellular communication, can encapsulate therapeutic agents and home tumors. However, PD-L1 overexpression in TDEVs leads to systemic immunosuppression during in vivo circulation, ultimately inhibiting intratumoral T activity. In this study, CRISPR/Cas9-edited Pd-l1KO TDEV-fusogenic anthracycline doxorubicin (DOX) liposomes with high drug encapsulation (97%) are fabricated, which homologously deliver DOX to breast cancer cells to intensify the immunogenic response and induce PD-L1 overexpression in the tumor. By setting the stage for sensitizing tumors to ICIs, sequential treatment with disulfide-linked PD1-cross-anchored TDEVs nanogels at one-day interval could sustainably release PD1 in the tumor, triggering a high proportion of effector T cell-mediated destruction of orthotopic and metastatic tumors without off-target side effects in the 4T1-bearing TNBC mouse model. Such a TDEV-tandem-augmented chemoimmunotherapeutic strategy with efficient cancer-homing delivery capacity and optimized ordinal-interval regime provides a solid foundation for developing chemoimmunotherapeutic formulations for TNBC therapy at the clinical level.

19.
J Control Release ; 352: 276-287, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36273531

RESUMO

Platinum (Pt)-based chemotherapy has been necessary for clinical cancer treatment. However, traditional bivalent drugs are hindered by poor physicochemical properties, severe toxic side effects, and drug resistance. Currently, elemental Pt(0) nanotherapeutics (NTs) have emerged to tackle the dilemma. The inherent acid-responsiveness of Pt(0) NTs could help to improve tumor selectivity and alleviate toxic effects. Moreover, the metal nature of Pt facilitates the great combination of Pt(0) NTs with photothermal and photodynamic therapy and imaging-guided diagnosis. Based on recent important researches, this review provides an updated introduction to Pt(0) NTs. First, the challenges of traditional Pt-based chemotherapy have been outlined. Then, Pt(0) NTs with multiple applications of tumor theranostics have been overviewed. Furthermore, the combinations of Pt(0) NTs with other therapeutical modalities are introduced. Last but not least, we envision the possible challenges and prospects associated with Pt(0) NTs.


Assuntos
Neoplasias , Fotoquimioterapia , Platina/uso terapêutico , Platina/química , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico
20.
Acta Pharm Sin B ; 12(12): 4348-4364, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36561998

RESUMO

Antibiotic resistance is one of the biggest threats to global health, as it can make the treatment of bacterial infections in humans difficult owing to their high incidence rate, mortality, and treatment costs. Bacteriophage, which constitutes a type of virus that can kill bacteria, is a promising alternative strategy against antibiotic-resistant bacterial infections. Although bacteriophage therapy was first used nearly a century ago, its development came to a standstill after introducing the antibiotics. Nowadays, with the rise in antibiotic resistance, bacteriophage therapy is in the spotlight again. As bacteriophage therapy is safe and has significant anti-bacterial activity, some specific types of bacteriophages (such as bacteriophage phiX174 and Pyo bacteriophage complex liquid) entered into phase III clinical trials. Herein, we review the key points of the antibiotic resistance crisis and illustrate the factors that support the renewal of bacteriophage applications. By summarizing recent state-of-the-art studies and clinical data on bacteriophage treatment, we introduced (i) the pharmacological mechanisms and advantages of antibacterial bacteriophages, (ii) bacteriophage preparations with clinical potential and bacteriophage-derived anti-bacterial treatment strategies, and (iii) bacteriophage therapeutics aimed at multiple infection types and infection-induced cancer treatments. Finally, we highlighted the challenges and critical perspectives of bacteriophage therapy for future clinical development.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa