Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Med ; 5(4): 348-367.e7, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38521069

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) cancer cells specifically produce abnormal oncogenic collagen to bind with integrin α3ß1 receptor and activate the downstream focal adhesion kinase (FAK), protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) signaling pathway. Collectively, this promotes immunosuppression and tumor proliferation and restricts the response rate of clinical cancer immunotherapies. METHODS: Here, by leveraging the hypoxia tropism and excellent motility of the probiotic Escherichia coli strain Nissle 1917 (ECN), we developed nanodrug-bacteria conjugates to penetrate the extracellular matrix (ECM) and shuttle the surface-conjugated protein cages composed of collagenases and anti-programmed death-ligand 1 (PD-L1) antibodies to PDAC tumor parenchyma. FINDINGS: We found the oncogenic collagen expression in human pancreatic cancer patients and demonstrated its interaction with integrin α3ß1. We proved that reactive oxygen species (ROS) in the microenvironment of PDAC triggered collagenase release to degrade oncogenic collagen and block integrin α3ß1-FAK signaling pathway, thus overcoming the immunosuppression and synergizing with anti-PD-L1 immunotherapy. CONCLUSIONS: Collectively, our study highlights the significance of oncogenic collagen in PDAC immunotherapy, and consequently, we developed a therapeutic strategy that can deplete oncogenic collagen to synergize with immune checkpoint blockade for enhanced PDAC treatment efficacy. FUNDING: This work was supported by the University of Wisconsin Carbone Cancer Center Research Collaborative and Pancreas Cancer Research Task Force, UWCCC Transdisciplinary Cancer Immunology-Immunotherapy Pilot Project, and the start-up package from the University of Wisconsin-Madison (to Q.H.).


Assuntos
Carcinoma Ductal Pancreático , Nanopartículas , Neoplasias Pancreáticas , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Integrina alfa3beta1 , Projetos Piloto , Neoplasias Pancreáticas/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Colágeno , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Microambiente Tumoral
2.
Adv Sci (Weinh) ; 11(16): e2309295, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38358998

RESUMO

Bacteria have distinctive properties that make them ideal for biomedical applications. They can self-propel, sense their surroundings, and be externally detected. Using bacteria as medical therapeutic agents or delivery platforms opens new possibilities for advanced diagnosis and therapies. Nano-drug delivery platforms have numerous advantages over traditional ones, such as high loading capacity, controlled drug release, and adaptable functionalities. Combining bacteria and nanotechnologies to create therapeutic agents or delivery platforms has gained increasing attention in recent years and shows promise for improved diagnosis and treatment of diseases. In this review, design principles of integrating nanoparticles with bacteria, bacteria-derived nano-sized vesicles, and their applications and future in advanced diagnosis and therapeutics are summarized.


Assuntos
Bactérias , Sistemas de Liberação de Medicamentos , Nanotecnologia , Humanos , Nanotecnologia/métodos , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/uso terapêutico
3.
Bioact Mater ; 35: 150-166, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38318228

RESUMO

Neutrophils have recently emerged as promising carriers for drug delivery due to their unique properties including rapid response toward inflammation, chemotaxis, and transmigration. When integrated with nanotechnology that has enormous advantages in improving treatment efficacy and reducing side effects, neutrophil-based nano-drug delivery systems have expanded the repertoire of nanoparticles employed in precise therapeutic interventions by either coating nanoparticles with their membranes, loading nanoparticles inside living cells, or engineering chimeric antigen receptor (CAR)-neutrophils. These neutrophil-inspired therapies have shown superior biocompatibility, targeting ability, and therapeutic robustness. In this review, we summarized the benefits of combining neutrophils and nanotechnologies, the design principles and underlying mechanisms, and various applications in disease treatments. The challenges and prospects for neutrophil-based drug delivery systems were also discussed.

4.
ACS Appl Bio Mater ; 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37905498

RESUMO

Gene therapy that can introduce, counteract, or replace genes possesses great potential to address diseases at their genetic roots. A wide range of technologies, such as RNA interference, genome editing, DNA transformation, and mRNA vaccines, have been extensively investigated to modulate gene expression in an attempt to treat a myriad of diseases. Despite the great promise of gene therapeutics, a series of intracellular and extracellular barriers must be surmounted, including rapid clearance in circulation, insufficient site-specific accumulation, suboptimal cellular internalization, and deficient transfection efficiency. Advances in the delivery systems for gene delivery bring about profound progress in enhancing the bioavailability and biocompatibility of gene therapeutics. Notably, bioinspired and biomimetic gene delivery systems have emerged, which draw inspiration from natural processes and recapitulate the desired traits and functions of viruses, bacteria, exosomes, and eukaryotic cells. The integration of bioinspired and biomimetic designs can overcome biological barriers, improve the pharmacokinetic profile, and efficiently transport gene therapeutics to target cells. As such, these platforms amplify the therapeutic efficacy and reduce side effects, thus expediting the clinical translation of gene therapy. Herein, we summarize the latest advances in designing bioinspired or biomimetic delivery systems, introduce their advantages, and discuss the obstacles to overcome with rational designs.

5.
AAPS J ; 25(5): 80, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37589825

RESUMO

Macrophages, as one of the most abundant tumor-infiltrating cells, play an important role in tumor development and metastasis. The frequency and polarization of tumor-associated macrophages (TAMs) correlate with disease progression, tumor metastasis, and resistance to various treatments. Pro-inflammatory M1 macrophages hold the potential to engulf tumor cells. In contrast, anti-inflammatory M2 macrophages, which are predominantly present in tumors, potentiate tumor progression and immune escape. Targeting macrophages to modulate the tumor immune microenvironment can ameliorate the tumor-associated immunosuppression and elicit an anti-tumor immune response. Strategies to repolarize TAMs, deplete TAMs, and block inhibitory signaling hold great potential in tumor therapy. Besides, biomimetic carriers based on macrophages have been extensively explored to prolong circulation, enhance tumor-targeted delivery, and reduce the immunogenicity of therapeutics to augment therapeutic efficacy. Moreover, the genetic engineering of macrophages with chimeric antigen receptor (CAR) allows them to recognize tumor antigens and perform tumor cell-specific phagocytosis. These strategies will expand the toolkit for treating tumors, especially for solid tumors, drug-resistant tumors, and metastatic tumors. Herein, we introduce the role of macrophages in tumor progression, summarize the recent advances in macrophage-centered anticancer therapy, and discuss their challenges as well as future applications. Graphical abstract.


Assuntos
Biomimética , Macrófagos , Humanos , Progressão da Doença , Engenharia Genética
6.
Adv Drug Deliv Rev ; 198: 114871, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37196699

RESUMO

The relay delivery strategy is a two-step targeting approach based on two distinct modules in which the first step with an initiator is to artificially create a target/environment which can be targeted by the follow-up effector. This relay delivery concept creates opportunities to amplify existing or create new targeted signals through deploying initiators to enhance the accumulation efficiency of the following effector at the disease site. As the "live" medicines, cell-based therapeutics possess inherent tissue/cell homing abilities and favorable feasibility of biological and chemical modifications, endowing them the great potential in specifically interacting with diverse biological environments. All these unique capabilities make cellular products great candidates that can serve as either initiators or effectors for relay delivery strategies. In this review, we survey recent advances in relay delivery strategies with a specific focus on the roles of various cells in developing relay delivery systems.


Assuntos
Sistemas de Liberação de Medicamentos , Humanos
7.
J Am Chem Soc ; 145(17): 9815-9824, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-37094179

RESUMO

Exploring the response of malignant cells to intracellular metabolic stress is critical for understanding pathologic processes and developing anticancer therapies. Herein, we developed ferritin-targeting proteolysis targeting chimeras (PROTACs) to establish the iron excess stress inside cancer cells and investigated subsequent cellular behaviors. We conjugated oleic acid that binds to the ferritin dimer to the ligand of von Hippel-Lindau (VHL) E3 ligase through an alkyl linker. The screened chimera, DeFer-2, degraded ferritin and then rapidly elevated the free iron content, thereby initiating the caspase 3-GSDME-mediated pyroptosis in cancer cells rather than typical ferroptosis that is always associated with iron ion overload. According to its structural and physicochemical characteristics, DeFer-2 was loaded into a tailored albumin-based nano-formulation, which substantially inhibited tumor growth and prolonged the survival time of mice bearing B16F10 subcutaneous tumors with negligible adverse effects. This study developed a ferritin-targeting PROTAC for iron overload stress, revealed iron metabolic dysregulation-mediated pyroptosis, and provided a PROTAC-based pyroptosis inducer for anticancer treatment.


Assuntos
Ferritinas , Proteína Supressora de Tumor Von Hippel-Lindau , Animais , Camundongos , Proteína Supressora de Tumor Von Hippel-Lindau/química , Ferritinas/metabolismo , Piroptose , Proteólise , Ferro/metabolismo
8.
Sci Adv ; 9(13): eadf6854, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36989364

RESUMO

Immune checkpoint inhibitors (ICIs) can reinvigorate T cells to eradicate tumor cells, showing great potential in combating various types of tumors. We propose a delivery strategy to enhance tumor-selective ICI accumulation, which leverages the responsiveness of platelets and platelet-derivatives to coagulation cascade signals. A fused protein tTF-RGD targets tumor angiogenic blood vessel endothelial cells and initiates the coagulation locoregionally at the tumor site, forming a "cellular hive" to recruit anti-PD-1 antibody (aPD-1)-conjugated platelets to the tumor site and subsequently activating platelets to release aPD-1 antibody to reactivate T cells for improved immunotherapy. Moreover, on a patient-derived xenograft breast cancer model, the platelet membrane-coated nanoparticles can also respond to the coagulation signals initiated by tTF-RGD, thus enhancing the accumulation and antitumor efficacy of the loaded chemotherapeutics. Our study illustrates a versatile platform technology to enhance the local accumulation of ICIs and chemodrugs by taking advantage of the responsiveness of platelets and platelet derivatives to thrombosis.


Assuntos
Neoplasias , Trombose , Animais , Humanos , Modelos Animais de Doenças , Células Endoteliais , Imunoterapia , Neoplasias/tratamento farmacológico , Oligopeptídeos , Trombose/tratamento farmacológico , Trombose/etiologia , Receptor de Morte Celular Programada 1/imunologia
9.
Chem Soc Rev ; 52(3): 1068-1102, 2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36633324

RESUMO

Cell-based therapy holds great potential to address unmet medical needs and revolutionize the healthcare industry, as demonstrated by several therapeutics such as CAR-T cell therapy and stem cell transplantation that have achieved great success clinically. Nevertheless, natural cells are often restricted by their unsatisfactory in vivo trafficking and lack of therapeutic payloads. Chemical engineering offers a cost-effective, easy-to-implement engineering tool that allows for strengthening the inherent favorable features of cells and confers them new functionalities. Moreover, in accordance with the trend of precision medicine, leveraging chemical engineering tools to tailor cells to accommodate patients individual needs has become important for the development of cell-based treatment modalities. This review presents a comprehensive summary of the currently available chemically engineered tools, introduces their application in advanced diagnosis and precision therapy, and discusses the current challenges and future opportunities.


Assuntos
Engenharia Celular , Medicina de Precisão , Humanos
10.
Adv Mater ; 35(15): e2210440, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36656162

RESUMO

Immunotherapy has achieved revolutionary success in clinics, but it remains challenging for treating hepatocellular carcinoma (HCC) characterized by high vascularization. Here, it is reported that metal-organic framework-801 (MOF-801) can be employed as a stimulator of interferon genes (STING) through Toll-like receptor 4 (TLR4) not just as a drug delivery carrier. Notably, cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODNs) and 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) STING agonist with vascular disrupting function coordinates with MOF-801 to self-assemble into a nanoparticle (MOF-CpG-DMXAA) that effectively delivers CpG ODNs and DMXAA to cells for synergistically improving the tumor microenvironment by reprogramming tumor-associated macrophages (TAMs), promoting dendritic cells (DCs) maturation, as well as destroying tumor blood vessels. In HCC-bearing mouse models, it is demonstrated that MOF-CpG-DMXAA triggers systemic immune activation and stimulates robust tumoricidal immunity, resulting in a superior immunotherapeutic efficiency in orthotopic and recurrent HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Estruturas Metalorgânicas , Camundongos , Animais , Estruturas Metalorgânicas/farmacologia , Proteínas de Membrana , Carcinoma Hepatocelular/terapia , Imunidade Inata , DNA , Microambiente Tumoral
11.
Sci Adv ; 8(45): eabp8798, 2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36367930

RESUMO

Inflammatory bowel diseases (IBDs) are often associated with elevated levels of reactive oxygen species (ROS) and highly dysregulated gut microbiota. In this study, we synthesized a polymer of hyaluronic acid-poly(propylene sulfide) (HA-PPS) and developed ROS-scavenging nanoparticles (HPN) that could effectively scavenge ROS. To achieve colon tissue targeting effects, the HPN nanoparticles were conjugated to the surface of modified probiotic Escherichia coli Nissle 1917 (EcN). To enhance the bacteriotherapy of EcN, we encapsulated EcN cells with a poly-norepinephrine (NE) layer that can protect EcN against environmental assaults to improve the viability of EcN in oral delivery and prolong the retention time of EcN in the intestine due to its strong mucoadhesive capability. In the dextran sulfate sodium-induced mouse colitis models, HPN-NE-EcN showed substantially enhanced prophylactic and therapeutic efficacy. Furthermore, the abundance and diversity of gut microbiota were increased after treatment with HPN-NE-EcN, contributing to the alleviation of IBDs.

12.
Nat Commun ; 13(1): 6321, 2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36280674

RESUMO

Pore-forming Gasdermin protein-induced pyroptosis in tumor cells promotes anti-tumor immune response through the release of pro-inflammatory cytokines and immunogenic substances after cell rupture. However, endosomal sorting complexes required for transport (ESCRT) III-mediated cell membrane repair significantly diminishes the tumor cell pyroptosis by repairing and subsequently removing gasdermin pores. Here, we show that blocking calcium influx-triggered ESCRT III-dependent membrane repair through a biodegradable nanoparticle-mediated sustained release of calcium chelator (EI-NP) strongly enhances the intracellularly delivered GSDMD-induced tumor pyroptosis via a bacteria-based delivery system (VNP-GD). An injectable hydrogel and a lyophilized hydrogel-based cell patch are developed for peritumoral administration for treating primary and metastatic tumors, and implantation for treating inoperable tumors respectively. The hydrogels, functioning as the local therapeutic reservoirs, can sustainedly release VNP-GD to effectively trigger tumor pyroptosis and EI-NP to prevent the ESCRT III-induced plasma membrane repair to boost the pyroptosis effects, working synergistically to augment the anti-tumor immune response.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Piroptose , Proteínas de Ligação a Fosfato/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Cálcio/metabolismo , Quelantes de Cálcio/metabolismo , Quelantes de Cálcio/farmacologia , Preparações de Ação Retardada/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Membrana Celular/metabolismo , Imunidade , Citocinas/metabolismo , Hidrogéis/metabolismo
13.
Adv Funct Mater ; 32(37)2022 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-36304724

RESUMO

Despite the rapid development of immunotherapy, low response rates, poor therapeutic outcomes and severe side effects still limit their implementation, making the augmentation of immunotherapy an important goal for current research. DNA, which has principally been recognized for its functions of encoding genetic information, has recently attracted research interest due to its emerging role in immune modulation. Inspired by the intrinsic DNA-sensing signaling that triggers the host defense in response to foreign DNA, DNA or nucleic acid-based immune stimulators have been used in the prevention and treatment of various diseases. Besides that, DNA vaccines allow the synthesis of target proteins in host cells, subsequently inducing recognition of these antigens to provoke immune responses. On this basis, researchers have designed numerous vehicles for DNA and nucleic acid delivery to regulate immune systems. Additionally, DNA nanostructures have also been implemented as vaccine delivery systems to elicit strong immune responses against pathogens and diseased cells. This review will introduce the mechanism of harnessing DNA-mediated immunity for the prevention and treatment of diseases, summarize recent progress, and envisage their future applications and challenges.

14.
Sci Transl Med ; 14(656): eabn1128, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35921473

RESUMO

Glioblastoma multiforme (GBM) remains incurable despite aggressive implementation of multimodal treatments after surgical debulking. Almost all patients with GBM relapse within a narrow margin around the initial resected lesion due to postsurgery residual glioma stem cells (GSCs). Tracking and eradicating postsurgery residual GSCs is critical for preventing postoperative relapse of this devastating disease, yet effective strategies remain elusive. Here, we report a cavity-injectable nanoporter-hydrogel superstructure that creates GSC-specific chimeric antigen receptor (CAR) macrophages/microglia (MΦs) surrounding the cavity to prevent GBM relapse. Specifically, we demonstrate that the CAR gene-laden nanoporter in the hydrogel can introduce GSC-targeted CAR genes into MΦ nuclei after intracavity delivery to generate CAR-MΦs in mouse models of GBM. These CAR-MΦs were able to seek and engulf GSCs and clear residual GSCs by stimulating an adaptive antitumor immune response in the tumor microenvironment and prevented postoperative glioma relapse by inducing long-term antitumor immunity in mice. In an orthotopic patient-derived glioblastoma humanized mouse model, the combined treatment with nanoporter-hydrogel superstructure and CD47 antibody increased the frequency of positive immune responding cells and suppressed the negative immune regulating cells, conferring a robust tumoricidal immunity surrounding the postsurgical cavity and inhibiting postoperative glioblastoma relapse. Therefore, our work establishes a locoregional treatment strategy for priming cancer stem cell-specific tumoricidal immunity with broad application in patients suffering from recurrent malignancies.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Receptores de Antígenos Quiméricos , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Glioblastoma/genética , Glioma/patologia , Glioma/terapia , Hidrogéis , Macrófagos/patologia , Camundongos , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Expert Opin Drug Deliv ; 19(10): 1337-1349, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35949105

RESUMO

INTRODUCTION: Cancer immunotherapies have created a new generation of therapeutics to employ the immune system to attack cancer cells. However, these therapies are typically based on biologics that are nonspecific and often exhibit poor tumor penetration and dose-limiting toxicities. Nanocarriers allow the opportunity to overcome these barriers as they have the capabilities to direct immunomodulating drugs to tumor sites via passive and active targeting, decreasing potential adverse effects from nonspecific targeting. In addition, nanocarriers can be multifunctionalized to deliver multiple cancer therapeutics in a single drug platform, offering synergistic potential from co-delivery approaches. AREAS COVERED: This review focuses on the delivery of cancer therapeutics using emerging nanocarriers to achieve synergistic results via co-delivery of immune-modulating components (i.e. chemotherapeutics, monoclonal antibodies, and genes). EXPERT OPINION: Nanocarrier-mediated delivery of combinatorial immunotherapy creates the opportunity to fine-tune drug release while achieving superior tumor targeting and tumor cell death, compared to free drug counterparts. As these nanoplatforms are constantly improved upon, combinatorial immunotherapy will afford the greatest benefit to treat an array of tumor types while inhibiting cancer evasion pathways.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia
16.
Chem Soc Rev ; 51(13): 5330-5350, 2022 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-35713468

RESUMO

Proteolysis Targeting Chimeras (PROTACs), an emerging therapeutic entity designed to degrade target proteins by hijacking the ubiquitin-proteasome system, have the potential to revolutionize the healthcare industry. The broad applicability of this protein degradation strategy has been verified with a few E3 ligases and a variety of distinct targets through the construction of modular chimeric structures. Despite recent efforts to promote the use of PROTACs for clinical applications, most PROTACs do not make it beyond the preclinical stage of drug development. There are several reasons that prevent PROTACs from reaching the market, and the inadequate delivery to the target site is one of the most challenging hurdles. With the increasing need for accelerating the translational process, combining the concepts of PROTACs and delivery systems has been explored to enhance the in vivo performance of PROTACs. These improved delivery strategies can eliminate unfavorable physicochemical properties of PROTACs, improve their targetability, and decrease their off-target side effects. The integration of powerful PROTACs and versatile delivery systems will inaugurate a burgeoning orientation for the field of targeted protein degradation. In this review, we will survey the latest progress in improving the in vivo degradation efficacy of PROTACs through delivery strategies, outline design principles for PROTAC-based delivery systems, discuss the current challenges with PROTACs, and outlook future opportunities in this field.


Assuntos
Descoberta de Drogas , Proteólise , Ubiquitina-Proteína Ligases
17.
J Mater Chem B ; 10(37): 7222-7238, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-35612089

RESUMO

With the outstanding achievement of chimeric antigen receptor (CAR)-T cell therapy in the clinic, cell-based medicines have attracted considerable attention for biomedical applications and thus generated encouraging progress. As the basic construction unit of organisms, cells harbor low immunogenicity, desirable compatibility, and a strong capability of crossing various biological barriers. However, there is still a long way to go to fix significant bottlenecks for their clinical translation, such as facile preparation, strict stability requirements, scale-up manufacturing, off-target toxicity, and affordability. The rapid development of biotechnology and engineering approaches in materials sciences has provided an ideal platform to assist cell-based therapeutics for wide application in disease treatments by overcoming these issues. Herein, we survey the most recent advances of various cells as bioactive ingredients and outline the roles of biomaterials in developing cell-based therapeutics. Besides, a perspective of cell therapies is offered with a particular focus on biomaterial-involved development of cell-based biopharmaceuticals.


Assuntos
Produtos Biológicos , Neoplasias , Receptores de Antígenos Quiméricos , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T , Linfócitos T
18.
Nat Commun ; 13(1): 1845, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35387972

RESUMO

Immunosuppressive cells residing in the tumor microenvironment, especially tumor associated macrophages (TAMs), hinder the infiltration and activation of T cells, limiting the anti-cancer outcomes of immune checkpoint blockade. Here, we report a biocompatible alginate-based hydrogel loaded with Pexidartinib (PLX)-encapsulated nanoparticles that gradually release PLX at the tumor site to block colony-stimulating factor 1 receptors (CSF1R) for depleting TAMs. The controlled TAM depletion creates a favorable milieu for facilitating local and systemic delivery of anti-programmed cell death protein 1 (aPD-1) antibody-conjugated platelets to inhibit post-surgery tumor recurrence. The tumor immunosuppressive microenvironment is also reprogrammed by TAM elimination, further promoting the infiltration of T cells into tumor tissues. Moreover, the inflammatory environment after surgery could trigger the activation of platelets to facilitate the release of aPD-1 accompanied with platelet-derived microparticles binding to PD-1 receptors for re-activating T cells. All these results collectively indicate that the immunotherapeutic efficacy against tumor recurrence of both local and systemic administration of aPD-1 antibody-conjugated platelets could be strengthened by local depletion of TAMs through the hydrogel reservoir.


Assuntos
Plaquetas , Micropartículas Derivadas de Células , Humanos , Hidrogéis , Imunoterapia/métodos , Recidiva Local de Neoplasia , Microambiente Tumoral , Macrófagos Associados a Tumor
19.
Natl Sci Rev ; 9(3): nwab172, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35265340

RESUMO

Chimeric antigen receptor T cell (CAR T) therapy was a milestone in the treatment of relapsed and refractory B cell malignancies. However, beneficial effects of CAR T cells have not been obtained in solid tumors yet. Herein, we implement a porous microneedle patch that accommodates CAR T cells and allows in situ penetration-mediated seeding of CAR T cells when implanted in the tumor bed or in the post-surgical resection cavity. CAR T cells loaded in the pores of the microneedle tips were readily escorted to the tumor in an evenly scattered manner without losing their activity. Such microneedle-mediated local delivery enhanced infiltration and immunostimulation of CAR T cells as compared to direct intratumoral injection. This tailorable patch offers a transformative platform for scattered seeding of living cells for treating a variety of tumors.

20.
Exploration (Beijing) ; 2(3): 20210106, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37323702

RESUMO

Immunotherapy strategies that use cell-based delivery systems have sparked much interest in the treatment of malignancies, owing to their high biocompatibility, excellent tumor targeting capability, and unique biofunctionalities in the tumor growth process. A variety of design principles for cell-based immunotherapy, including cell surface decoration, cell membrane coating, cell encapsulation, genetically engineered cell, and cell-derived exosomes, give cancer immunotherapy great potential to improve therapeutic efficacy and reduce adverse effects. However, the treatment efficacy of cell-based delivery methods for immunotherapy is still limited, and practical uses are hampered due to complex physiological and immunological obstacles, such as physical barriers to immune infiltration, immunosuppressive tumor microenvironment, upregulation of immunosuppressive pathways, and metabolic restriction. In this review, we present an overview of the design principles of cell-based delivery systems in cancer immunotherapy to maximize the therapeutic impact, along with anatomical, metabolic, and immunological impediments in using cell-based immunotherapy to treat cancer. Following that, a summary of novel delivery strategies that have been created to overcome these obstacles to cell-based immunotherapeutic delivery systems is provided. Also, the obstacles and prospects of next-step development of cell-based delivery systems for cancer immunotherapy are concluded in the end.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...